EP1981525B1 - Extended release of neuregulin for improved cardiac function - Google Patents

Extended release of neuregulin for improved cardiac function Download PDF

Info

Publication number
EP1981525B1
EP1981525B1 EP06840724.6A EP06840724A EP1981525B1 EP 1981525 B1 EP1981525 B1 EP 1981525B1 EP 06840724 A EP06840724 A EP 06840724A EP 1981525 B1 EP1981525 B1 EP 1981525B1
Authority
EP
European Patent Office
Prior art keywords
neuregulin
nrg
extended release
day
release
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
EP06840724.6A
Other languages
German (de)
French (fr)
Other versions
EP1981525A4 (en
EP1981525A1 (en
Inventor
Mingdong Zhou
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Zensun Shanghai Science and Technology Ltd
Original Assignee
Zensun Shanghai Science and Technology Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Zensun Shanghai Science and Technology Ltd filed Critical Zensun Shanghai Science and Technology Ltd
Priority to EP15151306.6A priority Critical patent/EP2918283B1/en
Priority to EP18153913.1A priority patent/EP3363455A1/en
Publication of EP1981525A1 publication Critical patent/EP1981525A1/en
Publication of EP1981525A4 publication Critical patent/EP1981525A4/en
Application granted granted Critical
Publication of EP1981525B1 publication Critical patent/EP1981525B1/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/18Growth factors; Growth regulators
    • A61K38/1883Neuregulins, e.g.. p185erbB2 ligands, glial growth factor, heregulin, ARIA, neu differentiation factor
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/56Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule
    • A61K47/59Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes
    • A61K47/60Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes the organic macromolecular compound being a polyoxyalkylene oligomer, polymer or dendrimer, e.g. PEG, PPG, PEO or polyglycerol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • A61P21/04Drugs for disorders of the muscular or neuromuscular system for myasthenia gravis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/18Antipsychotics, i.e. neuroleptics; Drugs for mania or schizophrenia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/16Otologicals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/04Inotropic agents, i.e. stimulants of cardiac contraction; Drugs for heart failure
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/127Liposomes

Definitions

  • This invention relates generally to compositions and methods for preventing, treating or delaying various cardiac diseases or disorders by extended release of neuregulin to a mammal.
  • Cardiac (ventricular) hypertrophy is an important adaptive physiological response to increased stress or demands for cardiac work.
  • One of the early cellular changes that occurs after a stimulus for hypertrophy is the synthesis of mitochondria and expansion of myofibrillar mass (wall thickening) with a proportional increase in the size of individual cells, but no (or minimal) increase in the number of cells.
  • hypertrophy The transition from hypertrophy to heart failure is characterized by several alterations in cellular organization.
  • normal hypertrophic cells have a large size with increased and well organized contractile units, as well as strong cell-cell adhesions.
  • pathologically hypertrophic cells which also have large size and accumulation of proteins, display disorganization of contractile proteins (disarray of sarcomeric structures) and poor cell-cell adhesions (disarray of myofibers).
  • contractile proteins disorganization of contractile proteins
  • myofibers poor cell-cell adhesions
  • ACE angiotensin-converting enzyme
  • ACE inhibitors have also been administered in combination with other drugs such as digitalis, which increases the force of the heart's contractions, and/or a diuretic, which helps relieve the heart's workload by causing the kidneys to remove more sodium and water from the bloodstream.
  • digitalis which increases the force of the heart's contractions
  • diuretic which helps relieve the heart's workload by causing the kidneys to remove more sodium and water from the bloodstream.
  • diuretics can improve some symptoms of heart failure but it is not suitable as a sole treatment.
  • NRGs comprise a family of structurally related growth and differentiation factors that include NRG1, NRG2, NRG3 and NRG4 and isoforms thereof.
  • NRG1 a family of structurally related growth and differentiation factors that include NRG1, NRG2, NRG3 and NRG4 and isoforms thereof.
  • NRG1 over 15 distinct isoforms of NRG1 have been identified and divided into two large groups, known as ⁇ - and ⁇ - types, on the basis of differences in the sequence of their essential epidermal growth factor (EGF)-like domains.
  • EGF essential epidermal growth factor
  • NRGs bind to the EGF receptor family, which comprises EGFR, ErbB2, ErbB3 and ErbB4, each of which plays an important role in multiple cellular functions, including cell growth, differentiation and survival. They are protein tyrosine kinase receptors, consisting of an extracellular ligand-binding domain, transmembrane domain and cytoplasmic tyrosine kinase domain. After NRG binds to the extracellular domain of ErbB3 or ErbB4, it induces a conformational change that leads to heterodimer formation between ErbB3, ErbB4 and ErbB2 or homodimer formation between ErbB4 itself, which results in phosphorylation of the receptors' C-terminal domain inside the cell membrane.
  • the phosphorylated intracellular domain then binds additional signal proteins inside the cell, activating the corresponding downstream AKT or ERK signaling pathway, and inducing a series of cell reactions, such as stimulation or depression of cell proliferation, cell differentiation, cell apoptosis, cell migration or cell adhesion.
  • these receptors mainly ErbB2 and ErbB4 are expressed in the heart.
  • NRG-1 ⁇ neuregulin-1 ⁇
  • ErbB2 The orphan receptor, ErbB2
  • ErbB3 The orphan receptor
  • ErbB3 can form heterodimer with ErbB3 or ErbB4 with higher affinity than ErbB3 or ErbB4 homodimers.
  • Research in neural development has indicated that the formation of the sympathetic nervous system requires an intact NRG-1 ⁇ , ErbB2 and ErbB3 signaling system. Targeted disruption of the NRG-1 ⁇ or ErbB2 or ErbB4 led to embryonic lethality due to cardiac development defects.
  • NRG-1 ⁇ has been shown to enhance sarcomere organization in adult cardiomyocytes.
  • the short-term administration of a recombinant NRG-1 ⁇ EGF-like domain significantly improves or protects against deterioration in myocardial performance in three distinct animal models of heart failure. More importantly, NRG-1 ⁇ significantly prolongs survival of heart failure animals, see WO 00/37095 .
  • Extended release of NRG greatly improves the effect of NRG in the treatment of heart failure and cardiac hypertrophy compared to NRG administered by other methods. Extended release of NRG also has the benefit of reducing the adverse side effects of NRG compared to NRG administered by other methods.
  • the invention provides NRG1 or a. functional fragment thereof for use in a method for preventing, treating or delaying heart failure in a mammal, the method comprising extended release of NRG into a mammal in need thereof, over a period of between 4 hours and 12 hours per day by means of an osmotic pump or a syringe pump.
  • the extended release of NRG into a mammal leads to sustained activation of the ERK signaling pathway in cardiac cells.
  • the extended release of NRG into a mammal results in sustained activation of the AKT signaling pathway in cardiac cells.
  • the extended release of NRG into a mammal enhances the EF and/or FS values of the left ventricle of mammal:
  • the extended release of NRG into a mammal prevents cardiac hypertrophy.
  • Extended release of NRG may be used for reducing the interior diameter of the left ventricle.
  • extended release of NRG into a mammal reduces the LVEDD value by greater than about 2%. More preferably, extended release of NRG into a mammal reduces the LVEDD value by greater than about 5%. Even more preferably, extended release of NRG into a mammal reduces the LVEDD value by greater than about 10%. More preferably, extended release of NRG into a mammal reduces the LVEDD value by greater than about 15%. Most preferably, extended release of NRG into a mammal reduces the LVEDD value by greater than about 20%.
  • extended release of NRG into a mammal reduces the LVESD value by greater than about 2%. More preferably, extended release of NRG into a mammal reduces the LVESD value by greater than about 5%. Even more preferably, extended release of NRG into a mammal reduces the LVESD value by greater than about 10%. Even more preferably, extended release of NRG into a mammal reduces 20 the LVESD value by greater than about 15%. Most preferably, extended release of NRG into a mammal decreases the LVESD value by greater than about 20%.
  • Extended release of NRG may further be used for causing cardiomyocyte growth and/or differentiation, wherein extended release of NRG into a mammal in need thereof activates the MAP kinase pathway in cardiac cells and causes growth and/or differentiation of the cardiomyocyte.
  • Extended release of NRG may also be used for inducing remodeling of muscle cell sarcomeric and cytoskeleton structures, or cell-cell adhesions, wherein extended release of NRG into a mammal in need thereof activates the MAP kinase pathway in cardiac cells and causes remodeling of the cell structures or the cell-cell adhesions.
  • Extended release of NRG may also be used for treating or preventing disassociation of cardiac muscle cell-cell adhesion and/or the disarray of sarcomeric structures in a mammal in need thereof.
  • the present disclosure also relates to compositions and methods for preventing, treating or delaying various diseases or disorders in mammals, particularly in humans, by extending the release of a NRG protein, or a functional fragment thereof, or a nucleic acid encoding a NRG protein, or a functional fragment thereof, or an agent that enhances production and/or function of said NRG.
  • diseases and disorders include generally those of the central and peripheral nervous system.
  • diseases and disorders include, various cardiovascular diseases, cancer, neural system disease and/or muscle diseases, including muscular dystrophy (e.g., Duchenne, Limb-girdle) and multiple sclerosis, spinal injury, eye and ear diseases, diabetes, schizophrenia, and Alzheimer's.
  • muscular dystrophy e.g., Duchenne, Limb-girdle
  • multiple sclerosis e.g., spinal injury, eye and ear diseases, diabetes, schizophrenia, and Alzheimer's.
  • the invention also relates to an extended release composition or formulation of NRG use in a method of preventing, treating or delaying heart failure in a mammal.
  • the composition or formulation sustains activation of the ERK signaling pathway in cardiac cells.
  • the composition or formulation sustains activation of the AKT signaling pathway in cardiac cells.
  • the composition or formulation enhances the EF and/or FS values of the mammal.
  • the composition or formulation prevents cardiac hypertrophy.
  • kits comprising a NRG composition or formulation and an extended release technology known in the art, including, but not limited to, an osmotic pump or syringe pump, poly-ethylene glycol (PEG) coupling, and/or liposome or microsphere packaging.
  • the kit may further comprises an instruction for using the NRG composition or formulation and or extended release technology in preventing, treating or delaying heart failure in a mammal; preventing, treating or delaying cardiac hypertrophy in a mammal; or reducing the interior diameter of the left ventricle in a mammal.
  • the present invention provides methods for treating or preventing heart failure in a mammal by extended release of a sustained or varied amount of NRG over a period of between 4 hours and 12 hours per day by means of an osmotic pump or a syringe pump.
  • the mammal is a human patient suffering from or at risk of developing heart failure.
  • neuregulin or “NRG” used in the present invention refers to proteins or peptides that can bind and activate ErbB2, ErbB3; ErbB4 or combinations thereof, including but not limited to all neuregulin isoforms, neuregulin EGF domain alone, polypeptides comprising neuregulin EGF-like domain, neuregulin mutants or derivatives, and any kind of neuregulin-like gene products that also activate the above receptors as described in detail below.
  • neuregulin used in the present invention binds to and activates ErbB2/ErbB4 or ErbB2/ErbB3 heterodimers.
  • Neuregulin also includes NRG-1, NRG-2, NRG-3, and NRG-4 proteins, peptides, fragments and compounds that mimic the activities of neuregulin.
  • Neuregulin used in the present invention can activate the above ErbB receptors and modulate their biological reactions, e.g., stimulate breast cancer cell differentiation and milk protein secretion; induce the differentiation of neural crest cell into Schwann cell; stimulate acetylcholine receptor synthesis in skeletal muscle cell; and/or improve cardiocyte differntiation, survival and DNA synthesis.
  • Neuregulin also includes those variants with conservative amino acid substitutions that do not substantially alter their biological activity.
  • Suitable conservative substitutions of amino acids are known to those of skill in this art and may be made generally without altering the biological activity of the resulting molecule. Those of skill in this art recognize that, in general, single amino acid substitutions in non-essential regions of a polypeptide do not substantially alter biological activity (see, e.g., Watson et al. Molecular Biology of the Gene, 4th Edition, 1987, The Bejacmin/Cummings Pub. co., p.224 ).
  • Neuregulin protein encompasses neuregulin protein and peptide.
  • Neuregulin nucleic acid encompasses neuregulin nucleic acid and neuregulin oligonucleotide.
  • EGF-like domain refers to a polypeptide motif encoded by the neuregulin gene that binds to and activates ErbB2, ErbB3, ErbB4, or combinations thereof, and bears a structural similarity to the EGF receptor-binding domain as disclosed in WO 00/64400 , Holmes et al., Science, 256:1205-1210 (1992 ); U.S. Patent Nos. 5,530,109 and 5,716,930 ; Hijazi et al., Int. J. Oncol., 13:1061-1067 (1998 ); Chang et al.
  • EGF-like domain binds to and activates ErbB2/ErbB4 or ErbB2/ErbB3 heterodimers.
  • EGF-like domain comprises the amino acid sequence of the receptor binding domain of NRG-1. In some embodiments, EGF-like domain comprises the amino acid sequence corresponding to amino acid residues 177-226, 177-237, or 177-240 of NRG-1.
  • EGF-like domain comprises the amino acid sequence of the receptor binding domain of NRG-2. In certain embodiments, EGF-like domain comprises the amino acid sequence of the receptor binding domain of NRG-3. In certain embodiments, EGF-like domain comprises the amino acid sequence of the receptor binding domain of NRG-4. In certain embodiments, EGF-like domain comprises the amino acid sequence of Ala Glu Lys Glu Lys Thr Phe Cys Val Asn Gly Gly Glu Cys Phe Met Val Lys Asp Leu Ser Asn Pro, as described in U.S. Pat. No. 5,834,229 .
  • an "effective amount" of an active agent for treating a particular disease is an amount that is sufficient to ameliorate, or in some manner reduce the symptoms associated with the disease.
  • the amount may cure the disease but, typically, is administered in order to ameliorate the symptoms of the disease.
  • active agent means any substance intended for the diagnosis, cure, mitigation, treatment, or prevention of disease in humans and other animals, or to otherwise enhance physical and mental well being.
  • amelioration of the symptoms of a particular disorder by administration of a particular active agent refers to any lessening, whether permanent or temporary, lasting or transient that can be attributed to or associated with administration of the agent.
  • treat refers to any manner in which the symptoms of a condition, disorder or disease are ameliorated or otherwise beneficially altered.
  • the effect may be prophylactic in terms of completely or partially preventing a disease or symptom thereof and/or may be therapeutic in terms of a partial or complete cure for a disease and/or adverse effect attributable to the disease.
  • Treatment also encompasses any pharmaceutical use of the compositions herein.
  • vector refers to discrete elements that are used to introduce heterologous DNA into cells for either expression or replication thereof. Selection and use of such vehicles are well known within the skill of the artisan.
  • An expression vector includes vectors capable of expressing DNA that are operatively linked with regulatory sequences, such as promoter, regions, that are capable of effecting expression of such DNA fragments.
  • an expression vector refers to a recombinant DNA or RNA construct, such as a plasmid; a phage, recombinant virus or other vector that, upon introduction into an appropriate host cell, results in expression of the cloned DNA.
  • Appropriate expression vectors are well known to those of skill in the art and include those that are replicable in eukaryotic cells and/or prokaryotic cells and those that remain episomal or those which integrate into the host cell genome.
  • cardiac muscle cell differentiation means a condition characterized by the decrease in DNA synthesis by more than 10%, inhibition of other factor-stimulated DNA synthesis more than 10%, well organized sarcomeric structures and cell-cell adhesions, sustained activation of MAP kinases, and enhanced expression of p21 CIPI . Further discussion is provided in WO00/37095 .
  • ejection fraction means the portion of blood that is pumped out of a filled ventricle as the result of a heartbeat. It may be defined by the following formula: (LV diastolic volume - LV systolic volume) / LV diastolic volume.
  • fractional shortening means a ratio of the change in the diameter of the left ventricle between the contracted and relaxed states. It may be defined by the following formula: (LV end diastolic diameter - LV end systolic diameter) / LV end diastolic diameter.
  • heart failure means an abnormality of cardiac function where the heart does not pump blood at the rate needed for the requirements of metabolizing tissues.
  • Heart failure includes a wide range of disease states such as congestive heart failure, myocardial infarction, tachyarrhythmia, familial hypertrophic cardiomyopathy, ischemic heart disease, idiopathic dilated cardiomyopathy, myocarditis and the like.
  • the heart failure can be caused by any number of factors, including, without limitation, ischemic, congenital, rheumatic, or idiopathic forms.
  • Chronic cardiac hypertrophy is a significantly diseased state which is a precursor to congestive heart failure and cardiac arrest.
  • myocardial infarction refers to a blockade of a coronary artery or blood flow interruption leading to focal necrosis of part of the myocardium caused by severe and persistent ischemia.
  • extended release refers to providing continuous therapeutic level of an active agent (e.g., neuregulin) over a period of time.
  • the extended release includes, without limitation various forms of release, such as continuous release, controlled release, delayed release, depot, gradual release, long-term release, programmed release, prolonged release, proportionate release, protracted release, repository, retard, slow release, spaced release, sustained release, time coat, timed release, delayed action, extended action, layered-time action, long acting, prolonged action; repeated action, slow acting, sustained action, sustained-action medications, and controlled release.
  • the ability to obtain extended release, controlled release, timed release, sustained release, delayed release, long acting, pulsatile delivery or immediate release is performed using well-known procedures and techniques available to the ordinarily skilled artisan.
  • the amount of time over which the active agent continues to be released depends on the characteristics of the active agent and the extended release technology or technologies used, but in all cases is longer than that of administration of the active agent without the extended release technology or technologies.
  • microsphere is synonymous with “microparticle”, “microcapsule”, “nanosphere”, “nanoparticle” and “nanocapsule” unless the context clearly dictates otherwise.
  • pegylate means to attach at least one Poly (ethylene glycol) molecule or at least one derivative of Poly (ethylene glycol) to an active agent or other molecule.
  • organized, or enhanced organization of sarcomeres or sarcomeric structures means a condition characterized by the straight array of contractile proteins revealed by immunofluorescent staining of ⁇ -actinin in cardiac muscle cells.
  • the straight array of ⁇ -actinin proteins in cells can be distinguished by microscopy and its connected photography.
  • disorganized or disarray of sarcomeres or sarcomeric structures means the opposite of the "organized, or enhanced organization of sarcomeres or sarcomeric structures"
  • organized, or enhanced organization of cytoskeleton structures means a condition characterized by the straight actin fibers revealed phalloidin staining of cardiac muscle cells.
  • the straight actin fibers in cells can be distinguished by microscopy and its connected photography as exampled in figures of this specification.
  • disorganized or disarray of cytoskeleton structures means the opposite of "organized, or enhanced organization of cytoskeleton structures”.
  • protein is synonymous with “polypeptide” or “peptide” unless the context clearly dictates otherwise.
  • sustained activation of MAP kinases means that the phosphorylated state of MAP kinases, p42/44, is maintained for at least 21 hr in cells. Further discussion is provided in WO00/3709 .
  • the drugs counteract each other and slow the development of ventricular muscle cell hypertrophy by less than 50% when administered together.
  • a synergistic effect is said to be obtained if the two drugs slow the development of ventricular muscle cell hypertrophy by more than 50% while not causing an unacceptable increase in adverse side effects.
  • cardiac hypertrophy means a condition characterized by an increase in the size of individual ventricular muscle cells, the increase in cell size being sufficient to result in a clinical diagnosis of the patient or sufficient as to allow the cells to be determined as larger (e.g., 2-fold or more larger than non-hypertrophic cells). It may be accompanied by accumulation of contractile proteins within the individual cardiac cells and activation of embryonic gene expression.
  • In vitro and in vivo methods for determining the presence of ventricular muscle cell hypertrophy are known.
  • non-exposed (normal) cells have a hypertrophy score of 3 and a surface area/cell of 581 ⁇ m2 and fully hypertrophied cells have a hypertrophy score of 7 and a surface area/cell of 1811 ⁇ m2, or approximately 200% of normal.
  • Cells with a hypertrophy score of 4 have a surface area/cell of 771 ⁇ m2, or approximately 30% greater size than non-exposed cells; cells with a hypertrophy score of 5 have a surface area/cell of 1109 ⁇ m2, or approximately 90% greater size than non-exposed cells; and cells with a hypertrophy score of 6 have a surface area/cell of 1366 ⁇ m2, or approximately 135% greater size than non-exposed cells.
  • the presence of ventricular muscle cell hypertrophy preferably includes cells exhibiting an increased size of about 15% (hypertrophy score 3.5) or more.
  • Inducers of hypertrophy vary in their ability to induce a maximal hypertrophic response as scored by the above-described assay. For example, the maximal increase in cell size induced by endothelin is approximately a hypertrophy score of 5.
  • suppression of cardiac hypertrophy means a reduction in one of the parameters indicating hypertrophy relative to the hypertrophic condition, or a prevention of an increase in one of the parameters indicating hypertrophy relative to the normal condition.
  • suppression of ventricular muscle cell hypertrophy can be measured as a reduction in cell size relative to the hypertrophic condition.
  • Suppression of ventricular muscle cell hypertrophy means a decrease of cell size of 10% or greater relative to that observed in the hypertrophic condition. More preferably, suppression of hypertrophy means a decrease in cell size of 30% or greater; most preferably, suppression of hypertrophy means a decrease of cell size of 50% or more.
  • hypertrophy score assay when phenylephrine is used as the inducing agent, these decreases would correlate with hypertrophy scores of about 6.5 or less, 5.0-5.5, and 4.0-5.0, respectively.
  • suppression is examined relative to the maximum cell size (or hypertrophic score) measured in the presence of that inducer.
  • Prevention of ventricular muscle cell hypertrophy is determined by preventing an increase in cell size relative to normal cells, in the presence of a concentration of inducer sufficient to fully induce hypertrophy.
  • prevention of hypertrophy means a cell size increase less than 200% greater than non-induced cells in the presence of maximally stimulating concentration of inducer. More preferably, prevention of hypertrophy means a cell size increase less than 135% greater than noninduced cells; and most preferably, prevention of hypertrophy means a cell size increase less than 90% greater than non-induced cells.
  • prevention of hypertrophy in the presence of a maximally-stimulating concentration of phenylephrine means a hypertrophic score of about 6.0-6.5, 5.0-5.5, and 4.0-4.5, respectively.
  • In vivo determination of hypertrophy may include measurement of cardiovascular parameters such as blood pressure, heart rate, systemic vascular resistance, contractility, force of heartbeat, concentric or dilated hypertrophy, left ventricular systolic pressure, left ventricular mean pressure, left ventricular end-diastolic pressure, cardiac output, stroke index, histological parameters, and ventricular size and wall thickness.
  • Animal models available for determination of development and suppression of ventricular muscle cell hypertrophy in vivo include the pressure-overload mouse model, RV murine dysfunctional model, transgenic mouse model, and post-myocardial infarction rat model. Medical methods for assessing the presence, development, and suppression of ventricular muscle cell hypertrophy in human patients are known, and include, for example, measurements of diastolic and systolic parameters, estimates of ventricular mass and pulmonary vein flows.
  • Hypertrophy may be from any cause which is responsive to retinoic acid, including congenital viral, idiopathic, cardiotrophic, or myotrophic causes, or as a result of ischemia or ischemic insults such as myocardial infarction.
  • the treatment is performed to stop or slow the progression of hypertrophy, especially after heart damage, such as from ischemia, has occurred.
  • the agent(s) is given immediately after the myocardial infarction, to prevent or lessen hypertrophy.
  • activity unit or "1U” means the quantity of standard product that can induce 50% maximal reaction.
  • the EC50 must be measured. For example, if the EC50 for a batch of product was 0.067 ⁇ g/ml then that would be one unit Further, if 1 ⁇ g of that product is being used then 14.93U (1 /0.067) is being used.
  • the EC50 can be determined by any method known in the art, including the method employed by the inventors in the Examples below. This determination of the activity unit is important for quality control of genetically engineered products and clinically used drugs, permits product from different pharmaceuticals and/or different batch numbers to be quantified with uniform criteria.
  • unit of neuregulin is determined by measuring the activity of neuregulin through kinase receptor activation enzyme-linked immunosorbant assay (KIRA-ELISA) as described in detail in Example 6 below and in WO03/099300 , and Sadick et al., 1996, Analytical Biochemistry, 235:207-14 , the contents of which are incorporated by reference in their entireties. Briefly, the assay measures neuregulin induced ErbB2 activation and phosphorylation on the adherent breast carcinoma cell line, MCF-7.
  • KIRA-ELISA kinase receptor activation enzyme-linked immunosorbant assay
  • Membrane proteins are solubilized via Triton X-100 lysis and the receptor is captured in ELISA wells coated with ErbB2-specific antibodies (e.g., H4) with no cross-reaction to ErbB3 or ErbB4. The degree of receptor phosphorylation is then quantified by antiphosphotyrosine ELISA.
  • ErbB2-specific antibodies e.g., H4
  • the present invention provides NRG for treating or preventing heart failure in a mammal by extended release of a sustained or varied amount of NRG as defined in the claims.
  • Any NRG e.g., NRG-1, NRG-2, NRG-3 and NRG-4 and isoforms thereof
  • protein, peptide or fragment can be used in the practice of this invention.
  • Neuregulin or NRG refers to proteins or peptides that can bind and activate ErbB2, ErbB3, ErbB4 or combinations thereof, including but not limited to all neuregulin isoforms, neuregulin EGF domain alone, polypeptides comprising neuregulin EGF-like-domain, neuregulin mutants or derivatives, and any kind of neuregulin-like gene products that also activate the above receptors as described in detail below.
  • neuregulin used in the present invention binds to and activate ErbB2/ErbB4 or ErbB2/ErbB3 heterodimers.
  • Neuregulin used in the present invention can activate the above ErbB receptors and modulate their biological reactions, e.g., stimulate breast cancer cell differentiation and milk protein secretion; induce the differentiation of neural crest cell into Schwann cell; stimulate acetylcholine receptor synthesis in skeletal muscle cell; and/or improve cardiocyte differentiation, survival and DNA synthesis.
  • Assays for measuring the receptor binding activity are known in the art. For example, cells transfected with ErbB-2 and ErbB-4 receptor can be used.
  • EC50 is measured by methods known in the art. EC50 is the concentration of ligands which can compete 50% of bound radiolabeled ligands off the receptor complex. The higher the EC50 value is, the lower the receptor binding affinity is.
  • Neuregulin includes any neuregulin and isoforms thereof known in the art, including but not limited to all isoforms of neuregulin-1 ("NRG-1"), neuregulin-1 ("NRG-2”), neuregulin-1 ("NRG-3”) and neuregulin-4 ("NRG-43”).
  • NRG-1 is described, for example, in U.S. Pat. Nos. 5,530,109 , 5,716,930 , and 7,037,888 ; Lemke, Mol. Cell. Neurosci.
  • NRG-2 is described, for example, in Chang et al., 1997, Nature 387:509-512 ; Carraway et al., 1997, Nature 387:512-516 ; Higashiyama et al., 1997, J. Biochem. 122:675-680 , Busfield et al., 1997, Mol. Cell. Biol. 17:4007-4014 and International Pat. Pub. No. WO 97/09425 ).
  • NRG-3 is described, for example, in Hijazi et al., 1998, Int. J. Oncol. 13:1061-1067 , the contents of which are incorported by reference in their entireties.
  • NRG-4 is described, for example, in Harari et al., 1999 Oncogene. 18:2681-89 .
  • Neuregulin also include neuregulin EGF domain alone, polypeptides comprising neuregulin EGF domain or neuregulin-like gene products that mimic the activities of neuregulin and binds and activates ErbB2, ErbB3, ErbB4 or combinations thereof.
  • epidermal growth factor-like domain or “EGF-like domain” refers to a polypeptide motif encoded by the neuregulin gene that binds to and activates ErbB2, ErbB3, ErbB4, or combinations thereof, and bears a structural similarity to the EGF receptor-binding domain as disclosed in WO 00/64400 , Holmes et al., Science, 256:1205-1210 (1992 ); U.S. Patent Nos.
  • Neuregulin used in the present invention comprises the EGF-like domain encoded by NRG-1.
  • EGF-like domain comprises the amino acid sequence of the receptor binding domain of NRG-1.
  • EGF-like domain comprises the amino acid sequence corresponding to amino acid residues 177-226,177-237, or 177-240 of NRG-1.
  • neuregulin used in the present invention comprises the amino acid sequence of :
  • neuregulin which comprises the EGF-like domain encoded by NRG-2 or the EGF-like domain encoded by NRG-3 or the EGF-like domain. encoded by NRG-4; or the amino acid sequence of Ala Glu Lys Glu Lys Thr Phe Cys Val Asn Gly Gly Glu Cys Phe Met Val Lys Asp Leu Ser Asn Pro, as described in U.S. Pat. No. 5,834,229 .
  • the present invention provides compositions for use in extended release of neuregulin for preventing, treating or delaying heart failure.
  • Extended release of neuregulin allows for simplification of administration scheme, improves clinical efficacy and attenuates adverse events, e.g., related to high blood level of neuregulin. It is contemplated that extended release of neuregulin over a certain period could induce or maintain expression of certain genes for cardiomyocyte growth and/or differentiation, remodeling of muscle cell sarcomeric and cytoskeleton structures, or cell-cell adhesions.
  • Extended release of neuregulin can be administered by any route according to the judgment of those of skill in the art, including but not limited to orally, inhalationally, parenterally (e.g., intravenously, intramuscularly, subcutaneously, or intradermally).
  • neuregulin is administered orally.
  • neuregulin is administered intravenously.
  • neuregulin is administered intramuscularly.
  • neuregulin is extendedly released to the bloodstream of a mammal.
  • Neuregulin can be administered by any extended release means or by any delivery devices that are known to those of ordinary skill in the art.
  • Such dosage forms can be used to provide extended release of neuregulin using, for example, hydropropylmethyl cellulose, other polymer matrices, gels, permeable membranes, osmotic systems, multilayer coatings, microparticles, liposomes, microspheres, or a combination thereof to provide the desired release profile in varying proportions.
  • single unit dosage forms suitable for oral administration such as, but not limited to, tablets, capsules, gelcaps, and caplets that are adapted for controlled-release.
  • Extended release of neuregulin provides continuous therapeutic level of neuregulin over a period of time.
  • neuregulin may be released over a period of 1 hour, 2 hours, 4 hours, 8 hours, 10 hours, 12 hours, 14 hours, 16 hours, 20 hours, 24 hours or longer.
  • neuregulin may be released over a period of 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days or longer.
  • neuregulin may be released over a period of 1 week, 2 weeks, 3 weeks, 4 weeks or longer.
  • neuregulin may be released over a period of 1 month, 2 months, 4 months, 8 months, 12 months or longer.
  • neuregulin may be released over a period of 1 years, 2 years, 3 years, 4 years or longer.
  • neuregulin is released over a period of between 1 hour and 2 week, between 2 hours and 2 week, between 4 hours to 24 hours, between 4 days and 10 days.
  • the amount of time over which neuregulin is released may depend on various factors such as the extended release technology or technologies used.
  • neuregulin is released over a period of between 4 hours and 12 hours per day by means of an osmotic pump or a syringe pump.
  • Extended release of neuregulin maintains neuregulin in the blood within a desirable range, particularly at a level which is at or above the minimum effective therapeutic level and is below the minimum toxic level over a period of time.
  • the serum concentration of neuregulin in patients who received an extended release neuregulin composition can be compared with serum concentrations of patients receiving a non-extended release neuregulin composition (e.g., intravenous administration) at a time when the maximum blood level concentration occurs (C max ).
  • the patients receiving an extended release neuregulin composition have a lower maximum serum concentration (C max ) of neuregulin than the patients receiving a non-extended neuregulin composition.
  • the patients receiving an extended release neuregulin composition have a C max less than about 90%, 80%, 70% or 60% of the C max in patients receiving a non-extended release neuregulin composition. More preferably, the patients receiving an extended release neuregulin composition have a C max less than about 50%, 40% or 30% of the C max in patients receiving a non-extended release neuregulin composition. Most preferably, the patients receiving an extended release neuregulin composition have a C max less than about 20%, 10% or less of the C max in patients receiving a non-extended release neuregulin composition.
  • a standard curve is drawn by radioactivity versus unlabeled neuregulin amount
  • concentration of neuregulin in the serum certain amount of serum is added to tube containing cells on ice, radiolabeled neuregulin (50,000cpm) is then added, and the sample solution is mixed and left at 4°C overnight. Next morning, cells are pelleted and the supernatant is sucked away before the radioactivity is counted. The radioactivity is counted and the amount of neuregulin in the serum can be calculated according to the standard curve.
  • Extended release profile means a release profile in which less than 50% of the total release of neuregulin that occurs over the course of implantation/insertion or other method of administering neuregulin in the body occurs within the first 24 hours of administration.
  • the extended release profile is selected from the group consisting of: (a) the 50% release point occurring at a time that is between 24 and 48 hours after implantation/insertion or other method of administration, (b) the 50% release point occurring at a time that is between 48 and 96 hours after implantation/insertion or other method of administration, (c) the 50% release point occurring at a time that is between 96 and 168 hours (1 week) after implantation/insertion or other method of administration, (d) the 50% release point occurring at a time that is between 1 and 2 weeks after implantation/insertion or other method of administration, (e) the 50% release point occurring at a time that is between 2 and 4 weeks after implantation/insertion or other method of administration, (f) the 50% release point occurring at a time that is between 4 and 8 weeks after implantation/insertion or other method of administration, (g) the 50% release point occurring at a time that is between 8 and 16 weeks after implantation/insertion or other method of administration, (h) the 50% release point occurring at a time that is between 16
  • DFL degree of fluctuation
  • the patients receiving an extended release composition have a DFL approximately 90%, 80%, 70% or 60% of the DFL in patients receiving a non-extended release composition. More preferably, the patients receiving an extended release composition have a DFL approximately 50%, 40%, or 30% of the DFL in patients receiving a non-extended release composition. Most preferably, the patients receiving an extended release neuregulin composition have a DFL approximately 20%, 10% or less of the DFL in patients receiving a non-extended release neuregulin composition.
  • the size and frequency of dosing is determined by the pharmacodynamic and pharmacokinetic properties of the active agent.
  • many active agents that are readily soluble in the body are usually absorbed rapidly and provide a sudden burst of available drug.
  • An example is hypotension patients taking rapid-release nifedipine products.
  • the use of an extended-release product avoids the high initial blood concentrations which cause the sudden reduction in blood pressure and other significant haemodynamic changes such as reflex tachycardia.
  • active agents are targeted and removed or destroyed by the body, e.g., immune system, proteases.
  • Drugs with short half-lives for this and other reasons often need to be given the active agent at frequent intervals to maintain blood concentrations within the therapeutic range.
  • the use of extended-release products may maintain therapeutic concentrations over prolonged periods.
  • a reduction in the number of daily doses offered by extended-release products has the potential to improve compliance.
  • specific extended release technologies are disclosed herein, the invention is more general than any specific extended release technology. This includes the discovery that extended release of NRG at low doses unexpectedly improves the function of infarct heart.
  • extended release drug delivery technologies there are numerous extended release drug delivery technologies currently known in the art. Several are generally discussed below as preferred extended release technologies, but they are offered solely for purposes of illustration and not limitation. Many other related and unrelated technologies are well known in the art and may be employed in the practice of the invention disclosed herein. Additionally, combinations of the extended release technologies discussed herein and/or other extended release technologies known in the art may be employed in the practice of this invention. For example, many companies with specific expertise in extended release drug delivery technologies - e.g., Alza Corp., Durect Corp., Gilead Sciences, Baxter Pharmaceuticals, Brookwood Pharmaceuticals and OctoPlus - offer products and services that can be employed in the practice of this invention. Additionally, a search of patents, published patent applications and related publications will provide those skilled in the art reading this disclosure with significant possible extended release technologies. Thus, one skilled in the art will be able to select the desired extended release technology or technologies for use in the practice of this invention.
  • the extended release of NRG into the blood comprises the use of an osmotic pump.
  • Osmotic devices have demonstrated utility in delivering beneficial active agents to a target area in a controlled manner over prolonged periods of time.
  • Known devices include tablets, pills, capsules and implantable devices. Tablets and pills can be taken orally, whereas other pumps are implanted subcutaneously or intraperitoneally, or attached to a catheter for intravenous, intracerebral or intra-arterial infusion.
  • a core is encased by a semipermeable membrane having at least one orifice.
  • the semipermeable membrane is permeable to water, but impermeable to the active agent.
  • water penetrates through the semipermeable membrane into the core containing osmotic excipients and the active agent Osmotic pressure increases within the core and the agent is displaced through the orifice at a controlled, predetermined rate.
  • the core contains more than one internal compartment.
  • a first compartment may contain the active agent.
  • a second compartment contains an osmotic agent and/or "driving member.” See, e.g., U.S. Pat. No. 5,573,776 , the contents of which are incorporated herein by reference.
  • This compartment may have a high osmolality, which causes water to flux into the pump through the semipermeable membrane. The influx of water compresses the first compartment. This can be accomplished, for example, by using a polymer in the second compartment, which swells on contact with the fluid. Accordingly, the agent is displaced at a predetermined rate.
  • the osmotic pump may comprise more than one active agent-containing compartment, with each compartment containing the same agent or a different agent
  • concentrations of the agent in each compartment, as well as the rate of release, may also be the same or different.
  • the rate of delivery is generally controlled by the water permeability of the semipermeable membrane.
  • the delivery profile of the pump is independent of the agent dispensed, and the molecular weight of an agent, or its physical and chemical properties, generally have no bearing on its rate of delivery. Further discussion regarding the principle of operation, the design criteria, and the delivery rate for osmotic pumps is provided in Theeuwes and Yum, Annals of Biomedical Engineering, Vol. 4, No. 4 (1976 ) and Urquhart et. al., Ann. Rev. Pharmacol. Toxicol. 24:199-236 (1984 ).
  • Osmotic pumps are well know in the art and readily available to one of ordinary skill in the art from companies experienced in providing osmotic pumps for extended release drug delivery.
  • ALZA's DUROS® technology is an implantable, nonbiodegradable, osmotically driven system that enables delivery of small drugs, peptides, proteins, DNA and other bioactive macromolecules for up to one year
  • ALZA's OROS® technology embodies tablets that employ osmosis to provide precise, controlled drug delivery for up to 24 hours
  • Osmotica Pharmaceutical's Osmodex® system includes a tablet, which may have more than one layer of the drug(s) with the same or different release profiles
  • Shire Laboratories' EnSoTrol® system solubilizes drugs within the core and delivers the solubilized drug through a laser-drilled hole by osmosis
  • Alzet ® Osmotic pumps are miniature, implantable pumps used for research in mice, rats and other laboratory animals.
  • Typical materials for the semipermeable membrane include semipermeable polymers known to the art as osmosis and reverse osmosis membranes, such as cellulose acylate, cellulose diacylate, cellulose triacylate, cellulose acetate, cellulose diacetate, cellulose triacetate, agar acetate, amylase triacetate, beta glucan acetate, acetaldehyde dimethyl acetate, cellulose acetate ethyl carbamate, polyamides, plyurethanes, sulfonated polystyrenes, cellulose acetate pphthalate, cellulose acetate methyl carbamate, cellulose acetate succinate, cellulose acetate dimethyl aminoacetate, cellulose acetate ethyl carbamate, cellulose acetate chloracetate, cellulose dipalmitate, cellulose dioctanoate, cellulose dicaprylate, cellulose dipentanlate, cellulose acetate valerate, cellulose
  • the osmotic agent(s) present in the pump may comprise any osmotically effective compound(s) that exhibit an osmotic pressure gradient across the semipermeable wall against the exterior fluid.
  • Effective agents include, without limitation, magnesium sulfate, calcium sulfate, magnesium chloride, sodium chloride, lithium chloride, potassium sulfate, sodium carbonate, sodium sulfite, lithium sulfate, potassium chloride, sodium sulfate, d-mannitol, urea, sorbitol, inositol, raffinose, sucrose, flucose, hydrophilic polymers such as cellulose polymers, mixtures thereof, and the like, as disclosed in U.S. Pat No. 6,713,086 .
  • the “driving member” is typically a hydrophilic polymer which interacts with biological fluids and swells or expands.
  • the polymer exhibits the ability to swell in water and retain a significant portion of the imbibed water within the polymer structure.
  • the polymers swell or expand to a very high degree, usually exhibiting a 2 to 50 fold volume increase.
  • the polymers can be non-crosslinked or crosslinked. Hydrophilic polymers suitable for the present purpose are well known in the art.
  • the orifice may comprise any means and methods suitable for releasing the active agent from the system.
  • the osmotic pump may include one or more apertures or orifices which have been bored through the semipermeable membrane by mechanical procedures known in the art, including, but not limited to, the use of lasers as disclosed in U.S. Pat. No 4,088,864 .
  • it may be formed by incorporating an erodible element, such as a gelatin plug, in the semipermeable membrane.
  • osmotic pumps Although specific embodiments of osmotic pumps are discussed above, the invention is more general than any specific extended release technology. This includes the discovery that extended release of NRG improves the function of infarct heart and reduces the interior diameter of the left ventricle. There are numerous variations and different types of osmotic pumps currently known in the art and may be employed in the practice of the invention disclosed herein.
  • the extended release of NRG into the blood comprises the coupling of the active agent to a polymer, such as Poly(ethylene glycol) (hereinafter referred to as "PEG").
  • PEG Poly(ethylene glycol)
  • Coupling PEG to biologically active agents has demonstrated utility in delivering active agents to a target area in a controlled manner over prolonged periods of time.
  • modification of proteins with PEG has been extensively used within the biotechnology industry to reduce the antigenicity of therapeutically active agents and to extend their in vivo availability.
  • coupling PEG to bovine adenosine deaminase using cyanuric chloride results in a loss of immunogenicity.
  • the PEG adduct of both human growth hormone and E. coli L-asparaginase has been shown to have an extended circulatory half-life.
  • Coupling PEG to an active agent or other molecules can improve the efficacy and half-life of the active agent or other molecule, and also reduce its toxicity.
  • the PEG molecule is hydrated and in rapid motion. This rapid motion causes the PEG to sweep out a large volume and prevents the approach and interference of other molecules, e.g., immune cells or proteases.
  • the PEG polymer chains can protect the attached molecule from immune response and other clearance mechanisms, sustaining availability of the active agent.
  • polyethylene glycol molecules are connected to the protein via a reactive group found on the protein.
  • Commonly amino groups such as those on lysine residues or at the N-terminus, are used for attachment.
  • U.S. Pat. Nos. 5,824,784 and 4,002,531 disclose such methods for attaching PEG to an enzyme by reductive alkylation. Lysine residues may be strategically substituted for other amino acids or inserted into a polypeptide sequence to provide additional points of attachment as disclosed in U.S. Pat: No. 4,904,584 . Additional methods are known in the art for attaching branched or "multi-armed" PEG-derivatives to proteins as disclosed in U.S. Pat. No. 5,932,462 .
  • PEGs can also be used to link macromolecules to a targeting ligand or moiety, which directs the macromolecules to particular areas of interest.
  • U.S. Pat. No. 6,436,386 discloses active agent-polymer conjugates attached to a hydroxyapatite-targeting moiety for delivery of the active agent, such as bone growth factors, to hydroxyapatite surfaces, such as bone.
  • PEG derivatives are both available and suitable for use in the preparation of PEG-conjugates.
  • NOF Corp.'s SUNBRIGHT® Series provides numerous PEG derivatives, including methoxypolyethylene glycols and activated PEG derivatives, such as methoxy-PEG amines, maleimides and carboxylic acids, for coupling by various methods to drugs; enzymes, phospholipids and other biomaterials and Nektar Therapeutics' Advanced PEGylation also offers diverse PEG-coupling technologies to improve the safety and efficacy of therapeutics.
  • PEG is a well known polymer having the properties of solubility in water and in many organic solvents, lack of toxicity, lack of immunogenecity, and also clear, colorless, odorless and stable.
  • One use of PEG is to covalently attach the polymer to insoluble molecules to make the resulting PEG-molecule conjugate soluble. For these reasons and others, PEG has been selected as the preferred polymer for attachment, but it has been employed solely for purposes of illustration and not limitation.
  • Similar products may be obtained with other water soluble polymers, including without limitation, poly(vinyl alcohol), other poly(alkylene oxides) such as poly(propylene glycol) and the like, poly(oxyethylated polyols) such as poly(oxyethylated glycerol) and the like, carboxymethylcellulose, dextran, polyvinyl alcohol, polyvinyl pyrrolidone, poly-1,3-dioxolane, poly-1,3,6-trioxane, ethylene/maleic anhydride, and polyaminoacids.
  • poly(vinyl alcohol) other poly(alkylene oxides) such as poly(propylene glycol) and the like
  • poly(oxyethylated polyols) such as poly(oxyethylated glycerol) and the like
  • carboxymethylcellulose carboxymethylcellulose
  • dextran dextran
  • polyvinyl alcohol polyvinyl pyrrolidone
  • the extended release of NRG into the blood comprises packaging NRG in a liposome, which has demonstrated utility in delivering beneficial active agents in a controlled manner over prolonged periods of time.
  • Liposomes are completely closed bilayer membranes containing an entrapped aqueous volume. Liposomes may be unilamellar vesicles possessing a single membrane bilayer or multilamellar vesicles with multiple membrane bilayers, each separated from the next by an aqueous layer.
  • the structure of the resulting membrane bilayer is such that the hydrophobic (non-polar) tails of the lipid orient toward the center of the bilayer while the hydrophilic (polar) heads orient towards the aqueous phase.
  • the active agent is entrapped in the liposome and then administered to the patient to be treated.
  • the active agent may associate with the lipid bilayer.
  • the immune system may recognize conventional liposomes as foreign bodies and destroy them before significant amounts of the active agent reaches the intended disease site.
  • the liposome may be coated with a flexible water soluble polymer that avoids uptake by the organs of the mononuclear phagocyte system, primarily the liver and spleen.
  • Suitable hydrophilic polymers for surrounding the liposomes include, without limitation, PEG, polyvinylpyrrolidone, polyvinylmethylether, polymethyloxazoline, polyethyloxazoline, polyhydroxypropyloxazoline, polyhydroxypropylmethacrylamide, polymethacrylamide, polydimethylacrylamide, polyhydroxypropylmethacrylate, polyhydroxethylacrylate, hydroxymethylcellulose hydroxyethylcellulose, polyethyleneglycol, polyaspartamide and hydrophilic peptide sequences as described in U.S. Pat. Nos. 6,316,024 ; 6,126,966 ; 6,056,973 ; 6,043,094 .
  • Liposomes may be comprised of any lipid or lipid combination known in the art.
  • the vesicle-forming lipids may be naturally-occurring or synthetic lipids, including phospholipids, such as phosphatidylcholine, phosphatidylethanolamine, phosphatidic acid, phosphatidylserine, phasphatidylglycerol, phosphatidylinositol, and sphingomyelin as disclosed in U.S. Patent Nos. 6,056,973 and 5,874,104 .
  • the vesicle-forming lipids may also be glycolipids, cerebrosides, or cationic lipids, such as 1,2-dioleyloxy-3-(trimethylamino) propane (DOTAP); N-[1-(2,3,-ditetradecyloxy)propyl]-N,N-dimethyl-N-hydroxyethylammonium bromide (DMRIE); N-[1[(2,3,-dioleyloxy)propyl]-N,N-dimethyl-N-hydroxy ethylammonium bromide (DORIE); N-[1-(2,3-dioleyloxy)propyl]-N,N,N-trimethylammonium chloride (DOTMA); 3 [N-(N',N'-dimethylaminoethane) carbamoly] cholesterol (DC-Chol); or dimethyldioctadecylammonium (DDAB) also as disclosed in U.S. Patent No. 6,056,97
  • the liposome may be targeted to specific sites within the body of a mammal by the attachment of a targeting ligand or moiety.
  • the targeting ligands are believed to be recognized by receptors or other compounds on the surface of target cells.
  • Typical target ligands include antibodies or antibody fragments, cell-receptor ligands, lectins and the like.
  • Such targeting ligands can be attached to liposomes by any means known in the art for the covalent or noncovalent attachment of such ligands to lipsomes.
  • polymer coated liposomes have been modified to achieve site specific delivery of active agents, by attaching a targeting ligand to either the polar head group residues of liposomal lipid components or the free ends of the polymer chains forming the surface coat on the liposomes as described in U.S. Patent No. 6,316,024 and 6,043,094 .
  • Such attachments may be accomplished by, for example, the coupling of proteins to liposomes through the use of a crosslinking agent having at least one maleimido group and an amine reductive function as described in U.S. Pat. No.
  • the liposomes can be manufactured by standard techniques known to those of skill in the art. For example, in one embodiment, as disclosed in U.S. Pat. No. 5,916,588 , a buffered solution of the active agent is prepared. Then a suitable lipid, such as hydrogenated soy phosphatidylcholine, and cholesterol, both in powdered form, are dissolved in chloroform or the like and dried by rotoevaporation. The lipid film thus formed is resupsended in diethyl ether or the like and placed in a flask, and sonicated in a water bath during addition of the buffered solution of the active agent.
  • a buffered solution of the active agent is prepared.
  • a suitable lipid such as hydrogenated soy phosphatidylcholine, and cholesterol, both in powdered form, are dissolved in chloroform or the like and dried by rotoevaporation.
  • the lipid film thus formed is resupsended in diethyl
  • the liposome can be produced by any method generally accepted in the art for making liposomes, including, without limitation, the methods of the above-cited documents.
  • Liposomes are also well known in the art and readily available from companies experienced in providing liposomes for extended release drug delivery.
  • ALZA's (formerly Sequus Pharmaceuticals') STEALTH® liposomal technology for intravenous drug delivery uses a polyethylene glycol coating on liposomes to evade recognition by the immune system
  • Gilead Sciences (formerly Nexstar's) liposomal technology was incorporated into AmBisome®, and FDA approved treatment for fungal infections
  • NOF Corp. offers a wide variety of GMP-grade phospholipids, phospholipids derivatives, and PEG-phospholipids under the tradenames COATSOME® and SUNBRIGHT®.
  • Microspheres have demonstrated utility in delivering beneficial active agents to a target area in a controlled manner over prolonged periods of time. Microspheres are generally biodegradable and can be used for subcutaneous, intramuscular and intravenous administration.
  • each microsphere is composed of an active agent and polymer molecules.
  • the active agent may be centrally located within a membrane formed by the polymer molecules, or, alternatively dispersed throughout the microsphere because the internal structure comprises a matrix of the active agent and a polymer excipient.
  • the outer surface of the microsphere is permeable to water, which allows aqueous fluids to enter the microsphere, as well as solubilized active agent and polymer to exit the microsphere.
  • the polymer membrane may comprise crosslinked polymers as disclosed in U.S. Patent No. 6,395,302 .
  • the active agent is essentially released when the polymer is degraded.
  • the pore size of the crosslinked polymers are larger than the size of the active agent, the active agent is at least partially released by diffusion.
  • Typical materials for the outer membrane include the following categories of polymers: (1) carbohydrate-based polymers, such as methylcellulose, carboxymethyl cellulose-based polymers, dextran, polydextrose, chitins, chitosan, and starch (including hetastarch), and derivatives thereof; (2) polyaliphatic alcohols such as polyethylene oxide and derivatives thereof including polyethylene glycol (PEG), PEG-acrylates, polyethyleneimine, polyvinyl acetate, and derivatives thereof; (3) poly(vinyl) polymers such as poly(vinyl) alcohol, poly(vinyl)pyrrolidone, poly(vinyl)phosphate, poly(vinyl)phosphonic acid, and derivatives thereof; (4) polyacrylic acids and derivatives thereof; (5) polyorganic acids, such as polymaleic acid, and derivatives thereof; (6) polyamino acids, such as polylysine, and poly-imino acids, such as polyi
  • the microspheres are attached to or coated with additional molecules.
  • additional molecules can facilitate targeting, enhance receptor mediation, and provide escape from endocytosis or destruction.
  • Typical molecules include phospholipids, receptors, antibodies, hormones and polysaccharides.
  • one or more cleavable molecules may be attached to the outer surface of microspheres to target it to a predetermined site. Then, under appropriate biological conditions, the molecule is cleaved causing release of the microsphere from the target.
  • the microspheres are manufactured by standard techniques. For example, in one embodiment, volume exclusion is performed by mixing the active agent in solution with a polymer or mixture of polymers in solution in the presence of an energy source for a sufficient amount of time to form particles as disclosed in U.S. Pat. No. 6,268,053 .
  • the pH of the solution is adjusted to a pH near the isoelectric point (pI) of the macromolecule.
  • an energy source such as heat, radiation, or ionization, alone or in combination with sonication, vortexing, mixing or stirring, to form microparticles.
  • the resulting microparticles are then separated from any unincorporated components present in the solution by physical separation methods well known to those skilled in the art and may then be washed.
  • Microspheres are well known and readily available to one of ordinary skill in the art from companies experienced in providing such technologies for extended release drug delivery.
  • Epic Therapeutics a subsidiary of Baxter Healthcare Corp., developed PROMAXX®, a protein-matrix drug delivery system that produces bioerodible protein microspheres in a totally water-based process
  • OctoPlus developed OctoDEX®, crosslinked dextran microspheres that release active ingredients based on bulk degradation of matrix rather than based on surface erosion
  • Brookwood Pharmaceuticals advertises the availability of its microparticle technologies for drug delivery.
  • the amount of neuregulin used in the present invention will vary with the nature and severity of the disease or condition, and the route by which the active ingredient is administered.
  • the frequency and dosage will also vary according to factors specific for each patient depending on the specific therapy (e.g., therapeutic or prophylactic agents).
  • administered the severity of the disorder, disease, or condition, the route of administration, as well as age, body, weight, response, and the past medical history of the patient.
  • Effective doses may be extrapolated from dose-response curves derived from in vitro or animal model test systems.
  • Exemplary doses of neuregulin include milligram or microgram amounts of neuregulin per kilogram of subject or sample weight (e.g. about 1 microgram per kilogram to about 500 milligrams per kilogram, about 100 micrograms per kilogram to about 5 milligrams per kilogram, or about 1 microgram per kilogram to about 50 micrograms per kilogram).
  • the dosage administered to a patient is typically 0.001 mg/kg to 15 mg/kg of the patient's body weight, based on weight of the active peptide.
  • the dosage administered to a patient is between 0.001 mg/kg and 15 mg/kg, 0.005 mg/kg and 10 mg/kg, 0.01 mg/kg and 5 mg/kg, 0.001 mg/kg and 4 mg/kg, 0.005 mg/kg and 3 mg/kg, 0.01 mg/kg and 2 mg/kg, 0.001 mg/kg and 1 mg/kg, 0.005 mg/kg and 0.5 mg/kg, 0.010 mg/kg and 0.2 mg/kg, 0.005 mg/kg and 0.050 mg/kg of the patient's body weight.
  • Exemplary doses of neuregulin also include unit (U) or unit amounts of neuregulin per kilogram of subject or sample weight (e.g. , about 1 U per kilogram to about 5000 U per kilogram, about 10 U micrograms per kilogram to about 1000 per kilogram, or about 100 U per kilogram to about 500 U per kilogram).
  • the dosage administered to a patient is typically 10 U/kg to 1000 U/kg of the patient's body weight, based on weight of the active peptide.
  • the dosage administered to a patient is between 1 U/kg and 10,000U/kg, 1 U/kg and 5000 U/kg, 10 U/kg and 5000 U/kg, 10 U/kg and 1000 U/kg, 50 U/kg and 2000 U/kg, 50 U/kg and 1000/kg, 50 U/kg and 500 U/kg, 100 U/kg and 1000 U/kg, 100 U/kg and 500 U/kg, 100 U/kg and 200 U/kg, of the patient's body weight.
  • the recommended daily dose range of neuregulin in the methods of the invention for the conditions described herein lie within the range of from about 0.001 mg to about 1000 mg per day.
  • a total daily dose range should be between 0.001 mg per day and 15 mg per day, 0.005 mg per day and 10 mg per day, 0.01 mg per day and 5 mg per day, 0.001 mg per day and 4 mg per day, 0.005 mg per day and 3 mg per day; 0.01 mg per day and 2 mg per day, 0.001 mg per day and 1 mg per day, 0.005 mg per day and 0.5 mg per day, 0.010 mg per day and 0.2 mg per day.
  • the therapy can be initiated at a lower dose, perhaps about 0.1 ⁇ g to about 1 ⁇ g, and increased if necessary up to about 20 ⁇ g mg to about 1000 ⁇ g per day as either a single dose or divided doses, depending on the patient's global response. It may be necessary to use dosages of the active ingredient outside the ranges disclosed herein in some cases, as will be apparent to those of ordinary skill in the art. Furthermore, it is noted that the clinician or treating physician will know how and when to interrupt, adjust, or terminate therapy in conjunction with individual patient response. In certain embodiments, neuregulin is administered in an amount of about 1 U/day to about 10,000 U/day. In some embodiments, it is administered in an amount of about 1 U/day to about 5000 U/day.
  • it is administered in an amount of about 10U/day to about 2000 U/day. In some embodiments, it is administered in an amount of about 10 U/day to about 1000 U/day. In some embodiments, it is administered in an amount of about 100 U/day to about 200 U/day.
  • Neuregulin can also be administered in a dosing schedule or "therapeutic cycle.” Daily dosage of neuregulin in the therapeutic cycle is described in detail above.
  • the therapeutic cycle can last 2 days, 5 days, 7 days, 10 days, two weeks, three weeks, four weeks, five weeks, or six weeks.
  • neuregulin is administered daily for each day of the therapeutic cycle. In certain embodiments, neuregulin is administered consecutively for three, four, five, six, seven, eight, nine, ten, eleven or twelve days in a therapeutic cycle.
  • neuregulin in a therapeutic cycle is administered on day 1 of the cycle and the cycle concludes with one or more days of no neuregulin administration In some embodiments, neuregulin is administered daily for 3, 5, 7, or 10 days followed by a resting period in a therapeutic cycle.
  • the present invention is useful in preventing heart failure in patients being treated with a drug that causes cardiac hypertrophy or heart failure, e.g., fludrocortisone acetate or herceptin.
  • NRG may be administered prior to, simultaneously with, or subsequent to a drug which causes such cardiac diseases.
  • NRG is administered in combination with an effective amount of a compound that acts to suppress a different hypertrophy induction pathway than NRG.
  • NRG is administered with such hypertrophy suppressors and/or additional components, without limitation, a cardiotrophic inhibitor such as a Ct-1 (cardiotrophin-1) antagonist, an ACE inhibitor, such as captopril (Capoten®), and/or human growth hormone and/or IGF-I (Insulin like growth factor I) in the case of congestive heart failure, or with another anti-hypertrophic, myocardiotrophic factor, anti-arrhythmic, or inotropic factor in the case of other types of heart failure or cardiac disorder.
  • a cardiotrophic inhibitor such as a Ct-1 (cardiotrophin-1) antagonist, an ACE inhibitor, such as captopril (Capoten®), and/or human growth hormone and/or IGF-I (Insulin like growth factor I) in the case of congestive heart failure, or with another anti-hypertrophic, myo
  • NRG is administered in combination with current therapeutic approaches for treatment of heart failure, including, without limitation, ACE inhibitors and other vasodilators, diuretics, digitalis preparations, beta blockers, blood thinners, angiotensin II receptor blockers, calcium channel blockers or potassium.
  • ACE inhibitors which prevent the conversion of angiotensin I to angiotensin II, are vasodilators that cause the blood vessels to expand, lowering the blood pressure and reducing the heart's workload.
  • Vasodilators suitable for use in embodiments of the present invention include, without limitation, the following drugs: quinapril (Accupril®), ramipril (Altace®), captopril (Capoten®), benazepril (Lotensin®), fosinopril (Monopril®), lisinopril (Prinivil® or Zestril®), enalapril (Vasotec®), moexipril (Univasc®), trandolapril, and perindopril.
  • vasodilators useful in the present invention, include, without limitation, isosorbide dinitrate (Isordil®), nesiritide (Natrecor®), hydralazine (Apresoline®), nitrates and minoxidil.
  • Diuretics cause the kidneys to remove sodium and water from the blood stream, reducing the heart's workload, and include, without limitation, the following drugs: hydrochlorothiazide (HydroDIURIL®), chlorothiazide (Diuril®), furosemide (Lasix®), bumetanide (Bumex®), spironolactone (Aldactone®), triamterene (Dyrenium®); metolazone (Zaroxolyn®), torsemide, indapamide, polythiazide, amiloride, and combination agents (Dyazide®).
  • Digitalis preparations increase the force of the heart's contractions and include, without limitation, digoxin (Lanoxin®) and digitoxin.
  • Beta blockers reduce the heart's tendency to beat faster and include, without limitation, the following drugs: carvedilol (Coreg®) metoprolol (Lopressor® or Toprol XL®, atenolol, bisoprolol, labetalol, propranolol, sotalol, pindolol, penbutolol, acebutolol, timolol, nadolol, and betaxolol.
  • drugs include carvedilol (Coreg®) metoprolol (Lopressor® or Toprol XL®, atenolol, bisoprolol, labetalol, propranolol, sotalol, pindolol, penbutolol, acebutolol, timolol, nadolol, and betaxolol.
  • Blood thinners for use in embodiments of the present invention include, without limitation, warfarin (Coumadin®) and heparin.
  • Embodiments of the present invention may also use angiotensin II receptor blockers, which, rather than lowering the levels of angiotensin II (as ACE inhibitors do), prevents angiotensin II from effecting the heart and blood vessels.
  • Angiotensin II receptor blockers suitable for use in the present invention include, without limitation, iosartan (Cozaar®); valsartan (Diovan®), irbesartan (Avapro®), candesartan, eprosartan, telmisartan, and olmesartan.
  • Calcium channel blockers are generally used to treat high blood pressure often associated with heart failure.
  • Calcium channel blockers suitable for use in the present invention include, without limitation, amlodipine (Norvasc®).
  • NRG can be administered with endothelin receptor antagonists, such as antibodies to the endothelin receptor, and peptides or other such small molecule antagonists; 3-adrenoreceptor antagonists such as carvedilol; x,-adrenoreceptor antagonists; anti-oxidants; compounds having multiple activities (e.g., 3-blocker/a-blocker/anti-oxidant); carvedilol-like compounds or combinations of compounds providing multiple functions found in carvedilol; growth hormone, etc.
  • endothelin receptor antagonists such as antibodies to the endothelin receptor, and peptides or other such small molecule antagonists
  • 3-adrenoreceptor antagonists such as carvedilol
  • x,-adrenoreceptor antagonists such as carvedilol
  • anti-oxidants such as carvedilol
  • compounds having multiple activities e.g., 3-blocker/a-blocker/anti-oxidant
  • Neuregulin agonists alone or in combination with other hypertrophy suppressor pathway agonists or with molecules that antagonize known hypertrophy induction pathways, are useful as drugs for in vivo treatment of mammals experiencing heart failure, so as to prevent or lessen heart failure effects.
  • Therapeutic formulations of agonist(s) for treating heart disorders are prepared for storage by mixing the agonist(s) having the desired degree of purity with optional physiologically acceptable carriers, excipients, or stabilizers ( Remington's Pharmaceutical Sciences, 16th edition, Oslo, A., Ed., 1980 ), in the form of lyophilized cake or aqueous solutions.
  • Acceptable carriers, excipients, or stabilizers are non-toxic to recipients at the dosages and concentrations employed, and include buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid; low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or immunoglobins; hydrophilic polymers such as olyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, arginine or lysine; monosaccharides, disaccharides, and other carbohydrates including glucose, mannose, or dextrins; chelating agents such as EDTA; sugar alcohols such as mannitol or sorbitol; salt-forming counter ions such as sodium; and/or non-ionic surfactants such as Tween, Pluronics, or polyethylene glycol (PEG).
  • buffers such as phosphate, citrate, and other organic acids
  • antioxidants including ascorbic
  • the antagonist(s) are also suitably linked to one of a variety of nonproteinaceous polymers, e.g., polyethylene glycol, polypropylene glycol, or polyalkylenes, in the manner set forth in US Patent Nos. 4,640,835 ; 4,496,689 ; 4,301,144 ; 4,670,417 ; 4,791,192 or 4,179,337 .
  • the amount of carrier used in a formulation may range from about 1 to 99%, preferably from about 80 to 99%, optimally between 90 and 99% by weight
  • the agonist(s) to be used for in vivo administration should be sterile. This is readily accomplished by methods known in the art, for example, by filtration through sterile filtration membranes, prior to or following lyophilization and reconstitution.
  • the agonist(s) ordinarily will be stored in lyophilized form or in solution.
  • Therapeutic agonist compositions generally are placed into a container having a sterile access port, for example, an intravenous solution bag or yial having a stopper pierceable by a hypodermic injection needle.
  • the agonist(s) administration is in a chronic fashion only, for example, one of the following routes: injection or infusion by intravenous, intraperitoneal, intracerebral, intramuscular, intraocular, intraarterial, or intralesional routes, orally or using sustained-release systems as noted above.
  • sustained-release preparations include. semipermeable matrices of solid hydrophobic polymers containing the protein, which matrices are in form of shaped articles, e.g., films, or microcapsules.
  • sustained-release matrices include polyesters, hydrogels (e.g., poly(2-hydroxyethylmethacrylate) as described by Langer et al. (1981) J. Biomed. Mater. Res. 15: 167-277 and Langer (1982) Chem. Tech.
  • degradable lactic acidglycolic acid copolymers such as the Lupron DepotTM (injectable microspheres composed of lactic acid-glycolic acid copolymer and leuprolide acetate), and poly-D-(-)-3-hydroxybutyric acid ( EP 133,988 ).
  • the agonist(s) also may be entrapped in microcapsules prepared, for example, by coacervation techniques or by interfacial polymerization (for example, hydroxymethylcellulose or gelatin-microcapsules and poly[methylmethacylate] microcapsules, respectively), in colloidal drug delivery systems (for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules), or in macroemulsions.
  • colloidal drug delivery systems for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules
  • Sustained-release agonist(s) compositions also include liposomally entrapped agonists(s).
  • Liposomes containing agonists(s) are prepared by methods known in the art, for example, those disclosed in DE 3,218,121 ; Epstein et al. (1985) Proc. Natl. Acad. Sci. USA 82: 3688-3692 ; Hwang et al. (1980) Proc. Natl. Acad. Sci. USA 77:4030-4034 : EP 52,322 ; EP 36,676 ; EP 88,046 ; EP 143,949 ; EP 142,641 ; Japanese patent application 83-118008 ; US Patent Nos. 4,485,045 and 4,544,545 ; and EP 102,324 .
  • a specific example of suitable sustained-release formulation is in EP 647,449 .
  • NRG is combined with or administered in concert with other agents for treating congestive heart failure, including ACE inhibitors (as discussed above), CT-1 inhibitors, human growth hormone, and/or IGF-I.
  • agents for treating congestive heart failure including ACE inhibitors (as discussed above), CT-1 inhibitors, human growth hormone, and/or IGF-I.
  • the effective amounts of such agents, if employed will be at the clinician's discretion. Dosage administration and adjustment are determined by methods known to those skilled in the art to achieve the best management of congestive heart failure and ideally takes into account use of diuretics or digitalis, and conditions such as hypotension and renal impairment. The dose will additionally depend on such factors as the type of drug used and the specific patient being treated.
  • the amount employed will be the same dose as that used if the drug were to be administered without agonist; however, lower doses may be employed depending on such factors as the presence of side-effects the condition being treated, the type of patient, and the type of agonists and drug, provided the total amount of agents provides an effective dose for the condition being treated.
  • kits for carrying out the therapeutic regiments of the invention comprise in one or more containers therapeutically effective amounts of NRG described herein, alone or in combination with other agents, in pharmaceutically acceptable form and in combination with an extended release technology as described herein. Instructions are optionally included for administration of the extended release NRG composition by a physician or by the patient.
  • the invention resides in the discovery that extended release of NRG activates the AKT or ERK signaling pathway as effectively as NRG delivered by other methods, and improves the function of infarct heart much more than NRG delivered by other methods.
  • NRG's interactions with ErbB receptors has been implicated in other diseases and disorders, e.g., diseases of the central and peripheral nervous system.
  • diseases and disorders include, various cardiovascular diseases, cancer, neural system disease and/or muscle diseases, including muscular dystrophy (e.g., Duchenne, Limb-girdle) and multiple sclerosis, spinal injury, eye and ear diseases, diabetes, schizophrenia, and Alzheimer's.
  • NRG intravenous
  • IM intramuscular
  • IVGTT IV glucose tolerance test
  • Wistar male rats (Shanghai Animal Center of Chinese Academy of Science), which weighed 180 ⁇ 20 grams, were numbered, weighed, and divided into groups. Each group contained three rats.
  • One group received IV injection of 4 ml/kg (volume/body weight) of vehicle (10mM Na 2 HPO 4 -NaH 2 PO4, 150mM NaCl, 0.2% human serum albumin (HSA), 5% mannitol, pH 6.0) as a control.
  • vehicle 10mM Na 2 HPO 4 -NaH 2 PO4, 150mM NaCl, 0.2% human serum albumin (HSA), 5% mannitol, pH 6.0
  • Rats were killed separately at 20min, 1hr, 2hr, 4hr and 6hr.
  • the left ventricles of each group of rats were cut into pieces in cold lysis buffer (50mM Tris pH 7.4, 5mM EDTA, 150mM NaCl, 1% Triton X-100, 2mM Na 2 VO 4 , 50mM NaF, 2mM PMSF, protease inhibitor cocktail (no EDTA, Roche)) after pooled, and washed with cold PBS.
  • the ventricles were then homogenized in ice water and centrifuged (Kendro Biofuge) at 12,000 rpm for 5 min at 4°C in 1.5ml Eppendorf tubes.
  • the supernatant was collected and spun one more time, then stored at -80°C. The samples were thawed and spun again before use.
  • the protein concentration of each sample was determined by BCA protein assay (Pierce BCA protein assay kit). A certain amount of each sample was mixed with 2X sample buffer (0.125M Tris ph 6.8, 20% glycerol, 4% SDS, 0.2M DTT, 0.012% bromophenol blue) and boiled for electrophoresis before transfer to PVDF membrane (Millipore).
  • AKT and ERK The phosphorylation of AKT and ERK, as well as the amount of AKT and ERK in each sample was detected with antibodies (ERK antibody and phosphorylated ERK antibody (Santa Cruz Biotechnology); AKT antibody and phosphorylated AKT antibody (Cell Signaling)).
  • Fig 1 The time course of phosphorylation of AKT and ERK in the left ventricle of normal rats when NRG was infused by each of these different methods is shown in Fig 1 .
  • NRG infused by IM, IV and IVGTT all activated sustained phosphorylation of ERK.
  • AKT phosphorylation induced by each method peaked at 20 min and decreased at 1hr, but increased again at 2 hr, where it maintained a high level from 4 hr to 6 hr.
  • NRG infused constantly is as effective as injection of NRG.
  • IVGTT infusion is a potential method for treating poor cardiac conditions.
  • osmotic pump is a way to deliver NRG constantly (as IVGTT)
  • Wistar male rats (Shanghai Animal Center of Chinese Academy of Science), which weighed 200 ⁇ 20g, were anesthetized by intraperitoneally injecting 100mg/kg (drug/body weight) of ketamine. The neck and chest were depilated and sanitized. An incision was made in the middle front neck to expose the tracheae. An 18G catheter overneedle was inserted into the tracheae between the 3rd and 5th cartilage of tracheae.
  • a plastic cannula was pushed into the trachea 1-2 cm and fixed to connect the Rodent Ventilator (SAR-830/P ventilator -Inspiratory flow rate, 1ml/100g/breath; Respiratory rate, 60 breaths/min).
  • SAR-830/P ventilator -Inspiratory flow rate 1ml/100g/breath; Respiratory rate, 60 breaths/min.
  • Another incision was made on the left front chest.
  • the skin was blunt dissected to expose the fourth and fifth rib, then the fourth rib was cut by elbowed mosquito forceps.
  • the ventilator (as described above) was linked to the cannula and turned on, and the heart was exposed to check the status of lung and heart.
  • the pericardium was rived off to identify the left atria and the pulmonary arterious cone after the heart was exteriorized through the incision.
  • the left ventricle anterior descending coronary artery between them was ligated tight with 6/0 medical suture before the heart was replaced into the thorax.
  • the thoracic wall was stitched.
  • the ventilator was blocked to full fill the lung.
  • the chest muscle and skin was stitched after the air in the thoracic cavity was gently squeezed out.
  • the ventilators were removed from the rats until constant spontaneous respiration resumed.
  • the cardiac function of the rats was then examined by echocardiography (Philips Sonos 7500 S4 probe) on the 14th day after ligation.
  • the rats with ejection fraction (hereinafter "EF") values from 30 to 50 percent were separated and grouped (15 rats per group).
  • Rats in the vehicle group received 0.4ml/100g (volume/body weight) of vehicle by IV injection. The vehicle was injected once a day for five days, stopped for two days, and then injected for another five days.
  • the IM and IV groups of rats received IM and IV injection of NRG, respectively (the amount of NRG was 149.3U/kg (protein/body weight), the volume was 0.4ml/100g).
  • the NRG was injected once a day for five days, stopped for two days, and then injected for another five days.
  • the IVGTT group had osmotic pumps (ALZET osmotic pump 2ML1) implanted on the fifth day after grouping.
  • Each pump contained 2 ml of NRG solution, which contained 933.1U of NRG (as a rat now weighed about 250g) and the infusion speed was about 18.7U/kg/h.
  • the maximum drug concentration compared to about 2.67U/kg by IV injection.
  • the steel tube was then inserted into one end of a 5cm PE60 tube.
  • the syringe needle was inserted into another end of the PE60 tube.
  • the plunger of syringe was pushed to add NRG solution to the flow moderator until it was full.
  • the long tube of the flow moderator was then inserted into the pump until its white flange attached to the pump.
  • the needle was drawn out of the flow moderator before soaking the pump in sterile 0.9% saline at 37°C overnight.
  • the rats were anesthetized by Ketamine (as described above). The area between neck and shoulder of the rats was depilated and sanitized. The body was covered with a piece of sterile wet cloth. An incision was then carefully made in the skin between the scapulae to locate and separate the external jugular vein. The distal end of the vein from the heart was ligated. A small hole was made by eye scissors on the wall of the external jugular vein and enlarged by microforceps. The PE60 tube connected to the osmotic pump was inserted 2cm into the vein through the hole. The proximal end of the vein from the heart was then bound with PE60 tube to fix the tube.
  • the distal end of the vein surrounding the PE60 tube was tied tight to further fix the tube.
  • a tunnel was formed by blunt separation of the skin from the incision to scapula.
  • a pocket was finally made on the back of the rat in the midscapular region by spreading the skin further.
  • the pump was slid through the tunnel into the pocket with the flow moderator pointing away from the incision.
  • the skin incision was then closed with a suture.
  • the rats were put back into the animal room after revival and were fed as usual.
  • IVGTT and IV The function of MI heart following NRG infusion by IVGTT and IV is shown in Table 1 below.
  • IVS "LVEDD” "PW”, “LVESD”, “EF”, "FS” and “CC” stand for interventricular septum, left ventricle end diastolic dimension, posterior wall thickness, left ventricle end systolic dimension, ejection fraction, fractional shortening and cardiac cycle, respectively.
  • EF and FS reflect the contractility of heart, especially for left ventricle.
  • EF end diastolic volume - end systolic volume / end diastolic volume
  • FS end diastolic dimension - end systolic dimension / end diastolic dimension
  • NRG infused by osmotic pump dramatically increased the cardiac function of MI rats compared to the IV group.
  • the EF value - a measurement of the heart's blood pumping efficiency that can be used to estimate the function of the left ventricle - in the IVGTT group was 59.18% higher than that of the vehicle group, and 34.81% higher than the IV group.
  • the FS value - also a way of measuring left ventricle performance - of the IVGTT group was 73.79% higher than that of the vehicle group, and 44.0% higher than the IV group.
  • NRG infused by osmotic pump not only greatly increased the cardiac function of MI rats compare with the IV group, but also reduced the interior diameter of the left ventricle.
  • the average Left Ventricle End Diastolic Dimension (hereinafter referred to as "LVEDD”) of the IVGTT group was 9.98% smaller than that of the vehicle group, and 6.03% smaller than the IV group.
  • the Left Ventricle End Systolic Dimension (hereinafter referred to as "LVESD”) of the IVGTT group was 21.37% smaller than that of the vehicle group, and 15.15% smaller than the IV group.
  • syringe pump is used for extended release of neuregulin in human patients.
  • Syringe pump can pump the solution continuously at certain speed into the bloodstream through a needle injected into the vein in rat tail.
  • Neuregulin was intravenously infused by syringe pump at different speed for different time per day into MI rats to better time period and speed for treatment.
  • Grouped MI rats was treated by intravenous injection of 4ml/kg (volume/body weight) vehicle everyday for 10 days (group A); or intravenous injection of 10 ⁇ g/kg neuregulin (2.5 ⁇ g/ml) everyday for 10 days (group B); or intravenous syringe pump infusion of neuregulin (0.625 ⁇ g/ml) at 1.25 ⁇ g/kg/h with 4 hours per day for 10 days (group C); or intravenous syringe pump infusion of neuregulin (1.25 ⁇ g/ml) at 2.5 ⁇ g/kg/h with 4 hours per day for 10 days (group D); or intravenous syringe pump infusion of neuregulin (0.625 ⁇ g/ml) at 0.625 ⁇ g/kg/h with 8 hours per day for 10 days (group E); or intravenous syringe pump infusion of neuregulin (1.25 ⁇ g/ml) at 1.25 ⁇ g/kg/h with 8 hours per day for 10 days (group F).
  • Echocardiography was then performed for all groups to examine the function of heart.
  • Table 2 echocardiography data for MI rats after intravenous syringe pump infusion (ISPI) or IV injection of NRG IVS LVEDD PW LVESD EF FS HR cm cm cm cm % % /min
  • a vehicle 0.057 ⁇ 0.003 0.947 ⁇ 0.041 0.142 ⁇ 0.013 0.811 ⁇ 0.047 34.5 ⁇ 3.3 14.4 ⁇ 1.6 418 ⁇ 51
  • B IV 0.060 ⁇ 0.005 0.924 ⁇ 0.060 0.164 ⁇ 0.016 0.770 ⁇ 0.057 41.5 ⁇ 2.6 17.8 ⁇ 1.6 332 ⁇ 52
  • D ISPI 2.5 ⁇ g/kg/h 4h/day 0.061 ⁇ 0.004 0.943 ⁇ 0.058 0.160 ⁇ 0.015 0.762 ⁇ 0.055 43.7 ⁇ 5.4 19.0 ⁇ 2.9 391 ⁇ 41
  • HR means heart rate
  • IV infusion was started at the same time as extended hypodermic infusion so the IV group was treated with NRG for 7 days.
  • the amount of NRG for the IV group was also changed to 223.95U/kg.
  • Table 3 shows that extended hypodermic infusion of NRG significantly increased the cardiac function of MI rats compared to the IV and vehicle groups. Compared to vehicle group, extended hypodermic infusion of NRG enhanced the EF value of MI hearts 42.77%, the FS value 51.75%. As discussed above, the EF and FS values are ways of measuring the heart's blood pumping efficiency and can be used to estimate the function of the left ventricle. Thus, these results show that extended release of NRG is much more effective than conventional IV injection for improving cardiac function.
  • Extended hypodermic infusion of NRG also reduced interior diameter of the left ventricle. Specifically, the LVEDD of MI hearts decreased 10.98% and the LVESD decreased 18.72% compare to vehicle group. IV injection of NRG in this experiment did not have an obvious effect on the cardiac function of MI heart compare to vehicle. The results show that extended hypodermic infusion of NRG can reduce left ventricular volume and mass, thereby improving left ventricular health and performance, which suggests that it may also be used as a treatment for heart failure.
  • Neuregulin was further infused by syringe pump at different speed for different time per day into MI rats.
  • Grouped MI rats was treated by intravenous injection of 4ml/kg (volume/body weight) vehicle everyday for 10 days (group A); or intravenous injection of 10 ⁇ g/kg neuregulin (2.5 ⁇ g/ml) everyday for 10 days (group B); or hypodermic injection (HI) of 10 ⁇ g/kg neuregulin (2.5 ⁇ g/ml) everyday for 10 days (group C); hypodermic syringe pump infusion of neuregulin (1.25 ⁇ g/ml) at 2.5 ⁇ g/kg/h with 4 hours per day for 10 days (group D); or hypodermic syringe pump infusion of neuregulin (1.11 ⁇ g/ml) at 1.67 ⁇ g/kg/h with 6 hours per day for 10 days (group E); or hypodermic syringe pump infusion of neuregulin (1.25 ⁇ g/ml) at 1.25 ⁇ g/kg/h with 8 hours per day for 10 days (group F).
  • Echocardiography was then performed for all groups to examine the function of heart.
  • Table 4 echocardiography data for MI rats after hypodermic syringe pump infusion (HSPI) or IV injection of NRG IVS LVEDD PW LVESD EF FS HR cm cm cm cm % % /min
  • a vehicle 0.060 ⁇ 0.007 0.906 ⁇ 0.107 0.151 ⁇ 0.027 0.757 ⁇ 0.130 39.3 ⁇ 10.8 16.9 ⁇ 6.1 388 ⁇ 33
  • B IV 0.063 ⁇ 0.004 0.812 ⁇ 0.045 0.159 ⁇ 0.010 0.726 ⁇ 0.047 43.4 ⁇ 2.8 18.8 ⁇ 1.4 383 ⁇ 33
  • C HI 0.063 ⁇ 0.003 0.909 ⁇ 0.054 0.163 ⁇ 0.011 0.744+0.048 42.1 ⁇ 3.7 18.1 ⁇ 1.9 390 ⁇ 40
  • D HSPI 2.5 ⁇ g/kg/h 4h/day 0.065 ⁇ 0.007 0.933 ⁇ 0.055 0.160 ⁇ 0.016 0.754 ⁇ 0.069 44.2 ⁇ 6.5 19.3 ⁇ 3.4 385 ⁇ 32
  • HR means heart rate
  • neuregulin by IV enhanced the EF value of MI rats by 10.43%
  • neuregulin by hypodermic injection enhanced the EF value of MI rats by 7.12%
  • neuregulin by hypodermic syringe pump infusion for 4h/day enhanced the EF value by 12.47%
  • neuregulin by hypodermic syringe pump infusion for 6h/day made the EF value jump to 22.90%
  • neuregulin by hypodermic syringe pump infusion for 8h/day also raised the EF value by 20.10%.
  • neuregulin by IV enhanced the FS value of MI rats by 11.24%
  • neuregulin by hypodermic injection enhanced the FS value of MI rats by 7.10%
  • neuregulin by hypodermic syringe pump infusion for 4h/day enhanced the FS value by 14.20%
  • neuregulin by hypodermic syringe pump infusion for 6h/day made the FS value jump to 26.63%
  • neuregulin by hypodermic syringe pump infusion for 8h/day also raised the FS value by 26.04%.
  • PEG mPEG-SPA-5000, NEKTAR
  • 10mM PBS pH 8.0
  • the mixture was then loaded onto a gel filtration column (S100, Pharmacia) to separate the components. Each peak fraction was collected and its sample was prepared for SDS-PAGE. After electrophoresis, the gel was stained by BaI 2 and Coomassie brilliant blue sequentially to detect PEG and NRG separately.
  • the mixture contains PEG monomer, NRG-monoPEG, NRG-diPEG and NRG-polyPEG.
  • the mixture was loaded onto a S100 gel filtration column, the components were well separated into NRG-polyPEG and NRG-diPEG (peak1), NRG-monoPEG and PEG (peak2).
  • MCF-7 cells was harvested, counted, pelleted and resuspended into DMEM (with 10% serum and 9 ⁇ g/ml insulin) at 5 ⁇ 10 4 cells/ml. 100 ⁇ l cell suspension was added to each well of 96 well plate and the plate was incubated at 37°C overnight. The cells were then washed 3 times with PBS and grew in serum free DMEM for another 24 hours.
  • ErbB2 antibody H4 (Zensun, anti-ErbB2 monoclonal antibody) was diluted to 6 ⁇ g/ml by coating buffer (50mM Na 2 CO 3 -NaHCO 3 , pH9.6), and added to 96 well plate 50 ⁇ l/well. The plate was left at 4°C overnight to coat with antibody.
  • DMEM was sucked away from the starved MCF-7 cells, and 100 ⁇ l serial dilutions of NRG, NRG-monoPEG or NRG-diPEG in DMEM was added to each well separately. DMEM was added to two wells as blank. The plate was incubated at 37°C for 20min. The cells were washed once with PBS before adding 100 ⁇ l/well lysis buffer (50mM Hepes, pH 8.0,150mM NaCl, 2mM sodium orthovanadate, 0.01% thimerosal, 1% Triton X-100 and one protease inhibitor cocktail tablet per 25ml solution) and lysing at 4°C for 30min. The plate was then shaken gently to completely lyse and remove cells from the plate and centrifugated at 15000rpm for 15min.
  • 100 ⁇ l/well lysis buffer 50mM Hepes, pH 8.0,150mM NaCl, 2mM sodium orthovanadate, 0.01% thime
  • the plate with coating antibody was washed five times with washing buffer (10mM PBS, pH7.4, 0.05% Tween 20) before adding 200 ⁇ l well of 5% nonfat milk in washing buffer.
  • the plate was incubated at 37°C for 2 hours before washed again 3 times with washing buffer.
  • HRP horseradish peroxidase
  • the EC50 of NRG, NRG-monoPEG and NRG-diPEG was shown in Table 5.
  • Table 5 EC50 of NRG, NRG-monoPEG and NRG-diPEG sample EC50 ( ⁇ g/ml) NRG 0.070 NRG-monoPEG 0.070 NRG-diPEG 0.098
  • NRG-1 ⁇ was administered intravenously by syringe pump to two groups of monkeys, each consisting of twenty four healthy rhesus monkeys (twelve male and twelve female, weighing about 5-7kg).
  • Group I was infused with NRG-1 ⁇ for twelve hours a day for fourteen days, at the speed of 1 ug/kg/hr. No Side effect was observed in this group.
  • Group II was infused for twenty four hours a day for fourteen days, at the speed of 1ug/kg/hr. In Group II, about 3-5 ml pericardial effusion in the heart of monkeys was observed.
  • NRG-1 ⁇ Two groups of healthy individuals were administered the same amount of NRG-1 ⁇ per day for 10 days. Eight individuals in Group I, were infused with NRG-1 ⁇ for four hours each day for ten days at speed of 0.3 ⁇ g/kg/hr. In this group, each individual on average experienced gastrointestinal disorder about two times during the ten-day period. Six individuals were infused with NRG-1 ⁇ for two hours each day for ten days at speed of 0.6 ⁇ g/kg/hr. In Group II, each individual on average experienced gastrointestinal disorder about five times during the ten-day period.
  • myocardial infarcted rats were infused with NRG-1 ⁇ and gene expression pattern in the left ventricle of these rats was analyzed by microarray. Compare with myocardial infarcted rats infused with vehicle, rats infused with NRG have different gene expression pattern.
  • thymosin beta like protein mRNA level increased 3.10 times; defensin beta.1 mRNA level increased 2.87 times; growth associated protein mRNA level increased 2.16 times; mRNA level of thymosin beta 4, Laminin gamma 1, myocardin, PI3K gamma regulatory subunit almost all doubled, while mRNA level of Elastin and PI3K gamma was nearly the same as before. It shows that neuregulin changes the expression level of various proteins in heart.

Description

    FIELD OF THE INVENTION
  • This invention relates generally to compositions and methods for preventing, treating or delaying various cardiac diseases or disorders by extended release of neuregulin to a mammal.
  • BACKGROUND OF THE INVENTION
  • Cardiac (ventricular) hypertrophy is an important adaptive physiological response to increased stress or demands for cardiac work. One of the early cellular changes that occurs after a stimulus for hypertrophy is the synthesis of mitochondria and expansion of myofibrillar mass (wall thickening) with a proportional increase in the size of individual cells, but no (or minimal) increase in the number of cells.
  • When the ventricle is stressed, the initial response is an increase in sarcomere length. This is followed by an increase in the total muscle mass. When the overload is severe; myocardial contractility becomes depressed. In its mildest form, this depression is manifested by a reduction in the velocity of shortening of unloaded myocardium or by a reduction in the rate of force development during isometric contraction. As myocardial contractility becomes further depressed, a more extensive reduction in the velocity of shortening of unloaded myocardium occurs, now accompanied by a decline in isometric force development and shortening. At this point, circulatory compensation may still be provided by cardiac dilation and an increase in muscle mass, which tend to maintain wall stress at normal levels. As contractility falls further, overt congestive heart failure, reflected in a depression of cardiac output and work and/or an elevation of ventricular end-diastolic volume and pressure at rest, supervenes.
  • The transition from hypertrophy to heart failure is characterized by several alterations in cellular organization. For example, normal hypertrophic cells have a large size with increased and well organized contractile units, as well as strong cell-cell adhesions. In contrast, pathologically hypertrophic cells, which also have large size and accumulation of proteins, display disorganization of contractile proteins (disarray of sarcomeric structures) and poor cell-cell adhesions (disarray of myofibers). Thus, in pathological hypertrophy, the increased size and accumulation of contractile proteins are associated with disorganized assembly of sarcomeric structures and a lack of robust cell-cell interactions.
  • Heart failure affects approximately five million Americans, and more than 550,000 new patients are diagnosed with the condition each year. Current drug therapy for heart failure is primarily directed to angiotensin-converting enzyme (ACE) inhibitors, which are vasodilators that cause blood vessels to expand, lowering blood pressure and reducing the heart's workload. While the percent reduction in mortality has been significant, the actual reduction in mortality with ACE inhibitors has averaged only 3%-4%, and there are several potential side effects.
  • ACE inhibitors have also been administered in combination with other drugs such as digitalis, which increases the force of the heart's contractions, and/or a diuretic, which helps relieve the heart's workload by causing the kidneys to remove more sodium and water from the bloodstream. However, at least one study demonstrated no difference in survival associated with the use of digitalis compared with placebo in patients with Class II-III heart failure. Additionally, diuretics can improve some symptoms of heart failure but it is not suitable as a sole treatment.
  • Additional limitations are associated with other options for preventing or treating heart failure. For example, heart transplantation is clearly more expensive and invasive than drug treatment, and it is further limited by the availability of donor hearts. Use of mechanical devices, such as biventricular pacemakers, are similarly invasive and expensive. Thus, there has been a need for new therapies given the deficiencies in current therapies.
  • One promising new therapy involves administration of neuregulin (hereinafter referred to as "NRG") to a patient suffering from or at risk of developing heart failure. NRGs comprise a family of structurally related growth and differentiation factors that include NRG1, NRG2, NRG3 and NRG4 and isoforms thereof. For example, over 15 distinct isoforms of NRG1 have been identified and divided into two large groups, known as α- and β- types, on the basis of differences in the sequence of their essential epidermal growth factor (EGF)-like domains.
  • NRGs bind to the EGF receptor family, which comprises EGFR, ErbB2, ErbB3 and ErbB4, each of which plays an important role in multiple cellular functions, including cell growth, differentiation and survival. They are protein tyrosine kinase receptors, consisting of an extracellular ligand-binding domain, transmembrane domain and cytoplasmic tyrosine kinase domain. After NRG binds to the extracellular domain of ErbB3 or ErbB4, it induces a conformational change that leads to heterodimer formation between ErbB3, ErbB4 and ErbB2 or homodimer formation between ErbB4 itself, which results in phosphorylation of the receptors' C-terminal domain inside the cell membrane. The phosphorylated intracellular domain then binds additional signal proteins inside the cell, activating the corresponding downstream AKT or ERK signaling pathway, and inducing a series of cell reactions, such as stimulation or depression of cell proliferation, cell differentiation, cell apoptosis, cell migration or cell adhesion. Among these receptors, mainly ErbB2 and ErbB4 are expressed in the heart.
  • It has been shown that the EGF-like domains of NRG1, ranging in size from 50 to 64-amino acids, are sufficient to bind to and activate these receptors. Previous studies have shown that neuregulin-1β (NRG-1β) can bind directly to ErbB3 and ErbB4 with high affinity. The orphan receptor, ErbB2, can form heterodimer with ErbB3 or ErbB4 with higher affinity than ErbB3 or ErbB4 homodimers. Research in neural development has indicated that the formation of the sympathetic nervous system requires an intact NRG-1β, ErbB2 and ErbB3 signaling system. Targeted disruption of the NRG-1β or ErbB2 or ErbB4 led to embryonic lethality due to cardiac development defects. Recent studies also highlighted the roles of NRG-1β, ErbB2 and ErbB4 in the cardiovascular development as well as in the maintenance of adult normal heart function. NRG-1β has been shown to enhance sarcomere organization in adult cardiomyocytes. The short-term administration of a recombinant NRG-1β EGF-like domain significantly improves or protects against deterioration in myocardial performance in three distinct animal models of heart failure. More importantly, NRG-1β significantly prolongs survival of heart failure animals, see WO 00/37095 . These effects make NRG-1β promising as a broad spectrum therapeutic or lead compound for heart failure due to a variety of common diseases. However, there is still a need for more effective methods of using NRG, which can be used in a clinical setting for the prevention, treatment or delaying of heart failure and/or cardiac hypertrophy.
  • SUMMARY OF THE INVENTION
  • Extended release of NRG greatly improves the effect of NRG in the treatment of heart failure and cardiac hypertrophy compared to NRG administered by other methods. Extended release of NRG also has the benefit of reducing the adverse side effects of NRG compared to NRG administered by other methods.
  • The invention provides NRG1 or a. functional fragment thereof for use in a method for preventing, treating or delaying heart failure in a mammal, the method comprising extended release of NRG into a mammal in need thereof, over a period of between 4 hours and 12 hours per day by means of an osmotic pump or a syringe pump.
  • In one embodiment of the method for preventing, treating or delaying heart failure in a mammal, the extended release of NRG into a mammal leads to sustained activation of the ERK signaling pathway in cardiac cells.
  • In another embodiment of the method for preventing, treating or delaying heart failure in a mammal in need thereof, the extended release of NRG into a mammal results in sustained activation of the AKT signaling pathway in cardiac cells.
  • In another embodiment of the method for preventing, treating or delaying heart failure in a mammal in need thereof, the extended release of NRG into a mammal enhances the EF and/or FS values of the left ventricle of mammal:
  • In another embodiment of the method for preventing, treating or delaying heart failure in a mammal in need thereof, the extended release of NRG into a mammal prevents cardiac hypertrophy.
  • Extended release of NRG may be used for reducing the interior diameter of the left ventricle. In one aspect, extended release of NRG into a mammal reduces the LVEDD value by greater than about 2%. More preferably, extended release of NRG into a mammal reduces the LVEDD value by greater than about 5%. Even more preferably, extended release of NRG into a mammal reduces the LVEDD value by greater than about 10%. More preferably, extended release of NRG into a mammal reduces the LVEDD value by greater than about 15%. Most preferably, extended release of NRG into a mammal reduces the LVEDD value by greater than about 20%.
  • In another preferred embodiment, extended release of NRG into a mammal reduces the LVESD value by greater than about 2%. More preferably, extended release of NRG into a mammal reduces the LVESD value by greater than about 5%. Even more preferably, extended release of NRG into a mammal reduces the LVESD value by greater than about 10%. Even more preferably, extended release of NRG into a mammal reduces 20 the LVESD value by greater than about 15%. Most preferably, extended release of NRG into a mammal decreases the LVESD value by greater than about 20%.
  • Extended release of NRG may further be used for causing cardiomyocyte growth and/or differentiation, wherein extended release of NRG into a mammal in need thereof activates the MAP kinase pathway in cardiac cells and causes growth and/or differentiation of the cardiomyocyte.
  • Extended release of NRG may also be used for inducing remodeling of muscle cell sarcomeric and cytoskeleton structures, or cell-cell adhesions, wherein extended release of NRG into a mammal in need thereof activates the MAP kinase pathway in cardiac cells and causes remodeling of the cell structures or the cell-cell adhesions.
  • Extended release of NRG may also be used for treating or preventing disassociation of cardiac muscle cell-cell adhesion and/or the disarray of sarcomeric structures in a mammal in need thereof.
  • Additionally, because NRG's interaction with ErbB receptors has been implicated in other diseases and disorders, extended release of NRG may also greatly improve the effect of NRG in the treatment of such other diseases and disorders compared to NRG administered by other methods. Thus, the present disclosure also relates to compositions and methods for preventing, treating or delaying various diseases or disorders in mammals, particularly in humans, by extending the release of a NRG protein, or a functional fragment thereof, or a nucleic acid encoding a NRG protein, or a functional fragment thereof, or an agent that enhances production and/or function of said NRG. Such diseases and disorders include generally those of the central and peripheral nervous system. Examples of other diseases and disorders, include, various cardiovascular diseases, cancer, neural system disease and/or muscle diseases, including muscular dystrophy (e.g., Duchenne, Limb-girdle) and multiple sclerosis, spinal injury, eye and ear diseases, diabetes, schizophrenia, and Alzheimer's.
  • The invention also relates to an extended release composition or formulation of NRG use in a method of preventing, treating or delaying heart failure in a mammal. In one embodiment, the composition or formulation sustains activation of the ERK signaling pathway in cardiac cells. In another embodiment, the composition or formulation sustains activation of the AKT signaling pathway in cardiac cells. In another embodiment, the composition or formulation enhances the EF and/or FS values of the mammal. In yet another embodiment, the composition or formulation prevents cardiac hypertrophy.
  • The application also discloses a kit comprising a NRG composition or formulation and an extended release technology known in the art, including, but not limited to, an osmotic pump or syringe pump, poly-ethylene glycol (PEG) coupling, and/or liposome or microsphere packaging. The kit may further comprises an instruction for using the NRG composition or formulation and or extended release technology in preventing, treating or delaying heart failure in a mammal; preventing, treating or delaying cardiac hypertrophy in a mammal; or reducing the interior diameter of the left ventricle in a mammal.
  • Those and other aspects, objects, advantages and features of the invention will be apparent to those persons skilled in the art upon reading the disclosure of the invention as more fully described below.
  • BRIEF DESCRIPTION OF THE DRAWING
    • Figure 1 shows the phosphorylation of AKT and ERK in the left ventricle of rats over time after NRG was infused by intramuscular injection, intravenous injection and intravenous glucose tolerance test infusion. "P-AKT," "P-ERK" and "NRG" mean phosphorylated AKT, phosphorylated ERK and neuregulin. "im," "iv," and "ivgtt" mean intramuscular injection, intravenous injection and intravenous glucose tolerance test, respectively.
    • Figure 2 shows gel stained by BaI2 to detect PEG. In the figure, "mixture" means the solution of PEG and NRG mixture after their reaction. "M", "peak1", "peak2" and "peak3" stand for protein marker and elution peak fraction 1, 2 and 3 of the mixture from the S100 column. "NRG-mono-PEG", "NRG-di-PEG" and "NRG-poly-PEG" mean NRG coupled to one PEG, two PEG and multiple (at least 3) PEG, respectively.
    • Figure 3 shows gel coomassie stained to detect NRG protein. The abbreviations are the same as in figure 2. In the M lane, the molecular weight for each band (from bottom to above) is 14.4kD, 20.1kD, 31.0kD, 43.0kD, 66.2kD and 97.4kD respectively.
    DETAILED DESCRIPTION OF THE INVENTION
  • The present invention provides methods for treating or preventing heart failure in a mammal by extended release of a sustained or varied amount of NRG over a period of between 4 hours and 12 hours per day by means of an osmotic pump or a syringe pump. Preferably, the mammal is a human patient suffering from or at risk of developing heart failure.
  • For clarity of disclosure, and not by way of limitation, the detailed description of the nvention hereinafter is divided into the subsections that follow.
  • A. Definitions
  • Unless defined otherwise, all technical and scientific terms used herein have the same meaning as is commonly understood by one of ordinary skill in the art to which this invention belongs. If a definition set forth in this section is contrary to or otherwise inconsistent with a definition set forth in the patents, applications, published applications and other publications referenced herein, the definition set forth in this section prevails over the definition in that reference.
  • As used herein, the singular forms "a", "an", and "the" mean "at least one" or "one or more" unless the context clearly dictates otherwise.
  • As used herein, "neuregulin" or "NRG" used in the present invention refers to proteins or peptides that can bind and activate ErbB2, ErbB3; ErbB4 or combinations thereof, including but not limited to all neuregulin isoforms, neuregulin EGF domain alone, polypeptides comprising neuregulin EGF-like domain, neuregulin mutants or derivatives, and any kind of neuregulin-like gene products that also activate the above receptors as described in detail below. In preferred embodiments, neuregulin used in the present invention binds to and activates ErbB2/ErbB4 or ErbB2/ErbB3 heterodimers. Neuregulin also includes NRG-1, NRG-2, NRG-3, and NRG-4 proteins, peptides, fragments and compounds that mimic the activities of neuregulin. Neuregulin used in the present invention can activate the above ErbB receptors and modulate their biological reactions, e.g., stimulate breast cancer cell differentiation and milk protein secretion; induce the differentiation of neural crest cell into Schwann cell; stimulate acetylcholine receptor synthesis in skeletal muscle cell; and/or improve cardiocyte differntiation, survival and DNA synthesis. Neuregulin also includes those variants with conservative amino acid substitutions that do not substantially alter their biological activity. Suitable conservative substitutions of amino acids are known to those of skill in this art and may be made generally without altering the biological activity of the resulting molecule. Those of skill in this art recognize that, in general, single amino acid substitutions in non-essential regions of a polypeptide do not substantially alter biological activity (see, e.g., Watson et al. Molecular Biology of the Gene, 4th Edition, 1987, The Bejacmin/Cummings Pub. co., p.224).
  • Neuregulin protein encompasses neuregulin protein and peptide. Neuregulin nucleic acid encompasses neuregulin nucleic acid and neuregulin oligonucleotide.
  • As used herein, "epidermal growth factor-like domain" or "EGF-like domain" refers to a polypeptide motif encoded by the neuregulin gene that binds to and activates ErbB2, ErbB3, ErbB4, or combinations thereof, and bears a structural similarity to the EGF receptor-binding domain as disclosed in WO 00/64400 , Holmes et al., Science, 256:1205-1210 (1992); U.S. Patent Nos. 5,530,109 and 5,716,930 ; Hijazi et al., Int. J. Oncol., 13:1061-1067 (1998); Chang et al. Nature, 387:509-512 (1997); Carraway et al., Nature, 387:512-516 (1997); Higashiyama et al., J. Biochem., 122:675-680 (1997); and WO 97/09425 . In certain embodiments, EGF-like domain binds to and activates ErbB2/ErbB4 or ErbB2/ErbB3 heterodimers. In certain embodiments, EGF-like domain comprises the amino acid sequence of the receptor binding domain of NRG-1. In some embodiments, EGF-like domain comprises the amino acid sequence corresponding to amino acid residues 177-226, 177-237, or 177-240 of NRG-1. In certain embodiments, EGF-like domain comprises the amino acid sequence of the receptor binding domain of NRG-2. In certain embodiments, EGF-like domain comprises the amino acid sequence of the receptor binding domain of NRG-3. In certain embodiments, EGF-like domain comprises the amino acid sequence of the receptor binding domain of NRG-4. In certain embodiments, EGF-like domain comprises the amino acid sequence of Ala Glu Lys Glu Lys Thr Phe Cys Val Asn Gly Gly Glu Cys Phe Met Val Lys Asp Leu Ser Asn Pro, as described in U.S. Pat. No. 5,834,229 .
  • As used herein, an "effective amount" of an active agent for treating a particular disease is an amount that is sufficient to ameliorate, or in some manner reduce the symptoms associated with the disease. The amount may cure the disease but, typically, is administered in order to ameliorate the symptoms of the disease.
  • As used herein, "active agent" means any substance intended for the diagnosis, cure, mitigation, treatment, or prevention of disease in humans and other animals, or to otherwise enhance physical and mental well being.
  • As used herein, "amelioration" of the symptoms of a particular disorder by administration of a particular active agent refers to any lessening, whether permanent or temporary, lasting or transient that can be attributed to or associated with administration of the agent.
  • As used herein, "treat", "treatment" and "treating" refer to any manner in which the symptoms of a condition, disorder or disease are ameliorated or otherwise beneficially altered. The effect may be prophylactic in terms of completely or partially preventing a disease or symptom thereof and/or may be therapeutic in terms of a partial or complete cure for a disease and/or adverse effect attributable to the disease. Treatment also encompasses any pharmaceutical use of the compositions herein.
  • As used herein, "vector (or plasmid)" refers to discrete elements that are used to introduce heterologous DNA into cells for either expression or replication thereof. Selection and use of such vehicles are well known within the skill of the artisan. An expression vector includes vectors capable of expressing DNA that are operatively linked with regulatory sequences, such as promoter, regions, that are capable of effecting expression of such DNA fragments. Thus, an expression vector refers to a recombinant DNA or RNA construct, such as a plasmid; a phage, recombinant virus or other vector that, upon introduction into an appropriate host cell, results in expression of the cloned DNA. Appropriate expression vectors are well known to those of skill in the art and include those that are replicable in eukaryotic cells and/or prokaryotic cells and those that remain episomal or those which integrate into the host cell genome.
  • As used herein, "cardiac muscle cell differentiation" means a condition characterized by the decrease in DNA synthesis by more than 10%, inhibition of other factor-stimulated DNA synthesis more than 10%, well organized sarcomeric structures and cell-cell adhesions, sustained activation of MAP kinases, and enhanced expression of p21CIPI. Further discussion is provided in WO00/37095 .
  • As used herein, "ejection fraction" or "EF" means the portion of blood that is pumped out of a filled ventricle as the result of a heartbeat. It may be defined by the following formula: (LV diastolic volume - LV systolic volume) / LV diastolic volume.
  • As used herein, "fractional shortening" or "FS" means a ratio of the change in the diameter of the left ventricle between the contracted and relaxed states. It may be defined by the following formula: (LV end diastolic diameter - LV end systolic diameter) / LV end diastolic diameter.
  • As used herein, "heart failure" means an abnormality of cardiac function where the heart does not pump blood at the rate needed for the requirements of metabolizing tissues. Heart failure includes a wide range of disease states such as congestive heart failure, myocardial infarction, tachyarrhythmia, familial hypertrophic cardiomyopathy, ischemic heart disease, idiopathic dilated cardiomyopathy, myocarditis and the like. The heart failure can be caused by any number of factors, including, without limitation, ischemic, congenital, rheumatic, or idiopathic forms. Chronic cardiac hypertrophy is a significantly diseased state which is a precursor to congestive heart failure and cardiac arrest.
  • As used herein, "myocardial infarction" refers to a blockade of a coronary artery or blood flow interruption leading to focal necrosis of part of the myocardium caused by severe and persistent ischemia.
  • As used herein, "extended release" refer s to providing continuous therapeutic level of an active agent (e.g., neuregulin) over a period of time. The extended release includes, without limitation various forms of release, such as continuous release, controlled release, delayed release, depot, gradual release, long-term release, programmed release, prolonged release, proportionate release, protracted release, repository, retard, slow release, spaced release, sustained release, time coat, timed release, delayed action, extended action, layered-time action, long acting, prolonged action; repeated action, slow acting, sustained action, sustained-action medications, and controlled release. The ability to obtain extended release, controlled release, timed release, sustained release, delayed release, long acting, pulsatile delivery or immediate release is performed using well-known procedures and techniques available to the ordinarily skilled artisan.
  • The amount of time over which the active agent continues to be released depends on the characteristics of the active agent and the extended release technology or technologies used, but in all cases is longer than that of administration of the active agent without the extended release technology or technologies.
  • As used herein, "microsphere" is synonymous with "microparticle", "microcapsule", "nanosphere", "nanoparticle" and "nanocapsule" unless the context clearly dictates otherwise.
  • As used herein, "pegylate" means to attach at least one Poly (ethylene glycol) molecule or at least one derivative of Poly (ethylene glycol) to an active agent or other molecule.
  • As used herein, "organized, or enhanced organization of sarcomeres or sarcomeric structures" means a condition characterized by the straight array of contractile proteins revealed by immunofluorescent staining of α-actinin in cardiac muscle cells. The straight array of α-actinin proteins in cells can be distinguished by microscopy and its connected photography. As used herein, "disorganized or disarray of sarcomeres or sarcomeric structures" means the opposite of the "organized, or enhanced organization of sarcomeres or sarcomeric structures"
  • As used herein, "organized, or enhanced organization of cytoskeleton structures" means a condition characterized by the straight actin fibers revealed phalloidin staining of cardiac muscle cells. The straight actin fibers in cells can be distinguished by microscopy and its connected photography as exampled in figures of this specification. As used herein, "disorganized or disarray of cytoskeleton structures" means the opposite of "organized, or enhanced organization of cytoskeleton structures".
  • As used herein, "protein" is synonymous with "polypeptide" or "peptide" unless the context clearly dictates otherwise.
  • As used herein, "sustained activation of MAP kinases" means that the phosphorylated state of MAP kinases, p42/44, is maintained for at least 21 hr in cells. Further discussion is provided in WO00/3709 .
  • The terms "synergistic, "synergistic effect" and like are used herein to describe improved treatment effects obtained by combining one or more therapeutic agents with one or more retinoic acid compounds. Although a synergistic effect in some fields is meant an effect which is more than additive (e.g., 1+1=3), in the field of medical therapy an additive (1+1=2) or less than additive (1+1=1.6) effect may be synergistic. For example, if each of two drugs were to inhibit the development of ventricular muscle cell hypertrophy by 50% if given individually, it would not be expected that the two drugs would be combined to completely stop the development of ventricular muscle cell hypertrophy. In many instances, due to unacceptable side effects, the two drugs cannot be administered together. In other instances, the drugs counteract each other and slow the development of ventricular muscle cell hypertrophy by less than 50% when administered together. Thus, a synergistic effect is said to be obtained if the two drugs slow the development of ventricular muscle cell hypertrophy by more than 50% while not causing an unacceptable increase in adverse side effects.
  • As used herein "cardiac hypertrophy" means a condition characterized by an increase in the size of individual ventricular muscle cells, the increase in cell size being sufficient to result in a clinical diagnosis of the patient or sufficient as to allow the cells to be determined as larger (e.g., 2-fold or more larger than non-hypertrophic cells). It may be accompanied by accumulation of contractile proteins within the individual cardiac cells and activation of embryonic gene expression.
  • In vitro and in vivo methods for determining the presence of ventricular muscle cell hypertrophy are known. In vitro assays for ventricular muscle cell hypertrophy include those methods described WO00/37095 , e.g., increased cell size and increased expression of atrial natriuretic factor (ANP). Changes in cell size are used in a scoring system to determine the extent of hypertrophy. These changes can be viewed with an inverted phase microscope, and the degree of hypertrophy scored with an arbitrary scale of 7 to 0, with 7 being fully hypertrophied cells, and 3 being non-stimulated cells. The 3 and 7 states may be seen in Simpson et al. (1982) Circulation Res. 51: 787=801, Figure 2, A and B, respectively. The correlation between hypertrophy score and cell surface area (µm2) has been determined to be linear (correlation coefficient = 0.99). In phenylephrine-induced hypertrophy, non-exposed (normal) cells have a hypertrophy score of 3 and a surface area/cell of 581 µm2 and fully hypertrophied cells have a hypertrophy score of 7 and a surface area/cell of 1811 µm2, or approximately 200% of normal. Cells with a hypertrophy score of 4 have a surface area/cell of 771 µm2, or approximately 30% greater size than non-exposed cells; cells with a hypertrophy score of 5 have a surface area/cell of 1109 µm2, or approximately 90% greater size than non-exposed cells; and cells with a hypertrophy score of 6 have a surface area/cell of 1366 µm2, or approximately 135% greater size than non-exposed cells. The presence of ventricular muscle cell hypertrophy preferably includes cells exhibiting an increased size of about 15% (hypertrophy score 3.5) or more. Inducers of hypertrophy vary in their ability to induce a maximal hypertrophic response as scored by the above-described assay. For example, the maximal increase in cell size induced by endothelin is approximately a hypertrophy score of 5.
  • As used herein, "suppression of cardiac hypertrophy" means a reduction in one of the parameters indicating hypertrophy relative to the hypertrophic condition, or a prevention of an increase in one of the parameters indicating hypertrophy relative to the normal condition. For example, suppression of ventricular muscle cell hypertrophy can be measured as a reduction in cell size relative to the hypertrophic condition. Suppression of ventricular muscle cell hypertrophy means a decrease of cell size of 10% or greater relative to that observed in the hypertrophic condition. More preferably, suppression of hypertrophy means a decrease in cell size of 30% or greater; most preferably, suppression of hypertrophy means a decrease of cell size of 50% or more. Relative to the hypertrophy score assay when phenylephrine is used as the inducing agent, these decreases would correlate with hypertrophy scores of about 6.5 or less, 5.0-5.5, and 4.0-5.0, respectively. When a different agent is used as the inducing agent, suppression is examined relative to the maximum cell size (or hypertrophic score) measured in the presence of that inducer.
  • Prevention of ventricular muscle cell hypertrophy is determined by preventing an increase in cell size relative to normal cells, in the presence of a concentration of inducer sufficient to fully induce hypertrophy. For example, prevention of hypertrophy means a cell size increase less than 200% greater than non-induced cells in the presence of maximally stimulating concentration of inducer. More preferably, prevention of hypertrophy means a cell size increase less than 135% greater than noninduced cells; and most preferably, prevention of hypertrophy means a cell size increase less than 90% greater than non-induced cells. Relative to the hypertrophy score assay when phenylephrine is used as the inducing agent, prevention of hypertrophy in the presence of a maximally-stimulating concentration of phenylephrine means a hypertrophic score of about 6.0-6.5, 5.0-5.5, and 4.0-4.5, respectively.
  • In vivo determination of hypertrophy may include measurement of cardiovascular parameters such as blood pressure, heart rate, systemic vascular resistance, contractility, force of heartbeat, concentric or dilated hypertrophy, left ventricular systolic pressure, left ventricular mean pressure, left ventricular end-diastolic pressure, cardiac output, stroke index, histological parameters, and ventricular size and wall thickness. Animal models available for determination of development and suppression of ventricular muscle cell hypertrophy in vivo include the pressure-overload mouse model, RV murine dysfunctional model, transgenic mouse model, and post-myocardial infarction rat model. Medical methods for assessing the presence, development, and suppression of ventricular muscle cell hypertrophy in human patients are known, and include, for example, measurements of diastolic and systolic parameters, estimates of ventricular mass and pulmonary vein flows.
  • Hypertrophy may be from any cause which is responsive to retinoic acid, including congenital viral, idiopathic, cardiotrophic, or myotrophic causes, or as a result of ischemia or ischemic insults such as myocardial infarction. Typically, the treatment is performed to stop or slow the progression of hypertrophy, especially after heart damage, such as from ischemia, has occurred. Preferably, for treatment of myocardial infarctions, the agent(s) is given immediately after the myocardial infarction, to prevent or lessen hypertrophy.
  • As used herein, "activity unit" or "1U" means the quantity of standard product that can induce 50% maximal reaction. In other words, to determine the activity unit for a given active agent, the EC50 must be measured. For example, if the EC50 for a batch of product was 0.067 µg/ml then that would be one unit Further, if 1 µg of that product is being used then 14.93U (1 /0.067) is being used. The EC50 can be determined by any method known in the art, including the method employed by the inventors in the Examples below. This determination of the activity unit is important for quality control of genetically engineered products and clinically used drugs, permits product from different pharmaceuticals and/or different batch numbers to be quantified with uniform criteria.
  • In certain embodiments, unit of neuregulin is determined by measuring the activity of neuregulin through kinase receptor activation enzyme-linked immunosorbant assay (KIRA-ELISA) as described in detail in Example 6 below and in WO03/099300 , and Sadick et al., 1996, Analytical Biochemistry, 235:207-14, the contents of which are incorporated by reference in their entireties. Briefly, the assay measures neuregulin induced ErbB2 activation and phosphorylation on the adherent breast carcinoma cell line, MCF-7. Membrane proteins are solubilized via Triton X-100 lysis and the receptor is captured in ELISA wells coated with ErbB2-specific antibodies (e.g., H4) with no cross-reaction to ErbB3 or ErbB4. The degree of receptor phosphorylation is then quantified by antiphosphotyrosine ELISA.
  • B. Neuregulin
  • The present invention provides NRG for treating or preventing heart failure in a mammal by extended release of a sustained or varied amount of NRG as defined in the claims. Any NRG (e.g., NRG-1, NRG-2, NRG-3 and NRG-4 and isoforms thereof) protein, peptide or fragment can be used in the practice of this invention.
  • Neuregulin or NRG refers to proteins or peptides that can bind and activate ErbB2, ErbB3, ErbB4 or combinations thereof, including but not limited to all neuregulin isoforms, neuregulin EGF domain alone, polypeptides comprising neuregulin EGF-like-domain, neuregulin mutants or derivatives, and any kind of neuregulin-like gene products that also activate the above receptors as described in detail below. In preferred embodiments, neuregulin used in the present invention binds to and activate ErbB2/ErbB4 or ErbB2/ErbB3 heterodimers. Neuregulin used in the present invention can activate the above ErbB receptors and modulate their biological reactions, e.g., stimulate breast cancer cell differentiation and milk protein secretion; induce the differentiation of neural crest cell into Schwann cell; stimulate acetylcholine receptor synthesis in skeletal muscle cell; and/or improve cardiocyte differentiation, survival and DNA synthesis. Assays for measuring the receptor binding activity are known in the art. For example, cells transfected with ErbB-2 and ErbB-4 receptor can be used. After receptor expressing cells are incubated with excess amount of radiolabeled neuregulin, the cells are pelleted and the solution containing unbound radiolabeled neuregulin is removed before unlabeled neuregulin solution is added to compete with radiolabeled neuregulin. EC50 is measured by methods known in the art. EC50 is the concentration of ligands which can compete 50% of bound radiolabeled ligands off the receptor complex. The higher the EC50 value is, the lower the receptor binding affinity is.
  • Neuregulin includes any neuregulin and isoforms thereof known in the art, including but not limited to all isoforms of neuregulin-1 ("NRG-1"), neuregulin-1 ("NRG-2"), neuregulin-1 ("NRG-3") and neuregulin-4 ("NRG-43"). NRG-1 is described, for example, in U.S. Pat. Nos. 5,530,109 , 5,716,930 , and 7,037,888 ; Lemke, Mol. Cell. Neurosci. 1996, 7:247-262; Peles and Yarden, 1993, BioEssays 15:815-824, 1993; Peles et al., 1992, Cell 69, 205-216; Wen et al., 1992, Cell 69, 559-572, 1992, Holmes et al., 1992, Science 256:1205-1210, Falls et al., 1993, Cell 72:801-815, Marchionni et al. 1993, Nature 362:312-8. NRG-2 is described, for example, in Chang et al., 1997, Nature 387:509-512; Carraway et al., 1997, Nature 387:512-516; Higashiyama et al., 1997, J. Biochem. 122:675-680, Busfield et al., 1997, Mol. Cell. Biol. 17:4007-4014 and International Pat. Pub. No. WO 97/09425 ). NRG-3 is described, for example, in Hijazi et al., 1998, Int. J. Oncol. 13:1061-1067, the contents of which are incorported by reference in their entireties. NRG-4 is described, for example, in Harari et al., 1999 Oncogene. 18:2681-89.
  • Neuregulin also include neuregulin EGF domain alone, polypeptides comprising neuregulin EGF domain or neuregulin-like gene products that mimic the activities of neuregulin and binds and activates ErbB2, ErbB3, ErbB4 or combinations thereof. As used herein, "epidermal growth factor-like domain" or "EGF-like domain" refers to a polypeptide motif encoded by the neuregulin gene that binds to and activates ErbB2, ErbB3, ErbB4, or combinations thereof, and bears a structural similarity to the EGF receptor-binding domain as disclosed in WO 00/64400 , Holmes et al., Science, 256:1205-1210 (1992); U.S. Patent Nos. 5,530,109 and 5,716,930 ; Hijazi et al., Int. J. Oncol., 13:1061-1067 (1998); Chang et al., Nature, 387:509-512 (1997); Carraway et al., Nature, 387:512-516 (1997); Higashiyama et al., J. Biochem., 122:675-680 (1997); and WO 97/09425 .
  • Neuregulin used in the present invention comprises the EGF-like domain encoded by NRG-1. In some embodiments, EGF-like domain comprises the amino acid sequence of the receptor binding domain of NRG-1. In some embodiments, EGF-like domain comprises the amino acid sequence corresponding to amino acid residues 177-226,177-237, or 177-240 of NRG-1.
  • In preferred embodiments, neuregulin used in the present invention comprises the amino acid sequence of :
    • Ser His Leu Val Lys Cys Ala Glu Lys Glu Lys Thr Phe Cys Val Asn Gly Gly Glu Cys Phe Met Val Lys Asp Leu Ser Asn Pro Ser Arg Tyr Leu Cys Lys Cys Pro Asn Glu Phe Thr Gly Asp Arg Lys Gln Asn Tyr Val Met Ala Ser Phe Tyr Lys Ala Glu Glu Leu Tyr Gln (SEQ ID NO:1), which corresponds to amino acids 177-237 of human NRG-1. The human nucleic acid sequence encoding the fragment is:
      • agccatcttg taaaatgtgc ggagaaggagaaaactttct gtgtgaatgg aggggagtgc ttcatggtga aagacctttc aaacccctcg agatacttgt gcaagtgccc aaatgagttt attggtgate gctgccaaaa ctacgtaatg gcgagcttct acaaggcgga ggagctgtac cag (SEQ ID NO:2).
  • Disclosed is neuregulin which comprises the EGF-like domain encoded by NRG-2 or the EGF-like domain encoded by NRG-3 or the EGF-like domain. encoded by NRG-4; or the amino acid sequence of Ala Glu Lys Glu Lys Thr Phe Cys Val Asn Gly Gly Glu Cys Phe Met Val Lys Asp Leu Ser Asn Pro, as described in U.S. Pat. No. 5,834,229 .
  • C. Extended Release Technology in General
  • The present invention provides compositions for use in extended release of neuregulin for preventing, treating or delaying heart failure. Extended release of neuregulin allows for simplification of administration scheme, improves clinical efficacy and attenuates adverse events, e.g., related to high blood level of neuregulin. It is contemplated that extended release of neuregulin over a certain period could induce or maintain expression of certain genes for cardiomyocyte growth and/or differentiation, remodeling of muscle cell sarcomeric and cytoskeleton structures, or cell-cell adhesions.
  • Extended release of neuregulin can be administered by any route according to the judgment of those of skill in the art, including but not limited to orally, inhalationally, parenterally (e.g., intravenously, intramuscularly, subcutaneously, or intradermally). In certain embodiments, neuregulin is administered orally. In certain embodiments, neuregulin is administered intravenously. In certain embodiments, neuregulin is administered intramuscularly. In preferred embodiments, neuregulin is extendedly released to the bloodstream of a mammal.
  • Disclosed is that Neuregulin can be administered by any extended release means or by any delivery devices that are known to those of ordinary skill in the art. Specifically, any extended means or delivery devices for deliverying peptides described in U.S. Patent Nos.: 3,845,770 ; 3,916,899 ; 3,536,809 ; 3,598,123 ; and 4,008,719 , 5,674,533 , 5,059,595 , 5,591,767 , 5,120,548 , 5,073,543 , 5,639,476 , 5,354,556 , 5,639,480 , 5,733,566 , 5,739,108 , 5,891,474 , 5,922,356 , 5,972,891 , 5,984,945 , 5,993,855 , 6,045,830 , 6,087,324 , 6,113,943 , 6,197,350 , 6,248,363 , 6,264,970 , 6,267,981 , 6,376,461 , 6,419,961 , 6,589,548 , 6,613,358 , 6,699,500 , 6,740,634 , 6,838,076 , 6,866,866 , 7,087,246 . Such dosage forms can be used to provide extended release of neuregulin using, for example, hydropropylmethyl cellulose, other polymer matrices, gels, permeable membranes, osmotic systems, multilayer coatings, microparticles, liposomes, microspheres, or a combination thereof to provide the desired release profile in varying proportions. Further disclosed are single unit dosage forms suitable for oral administration such as, but not limited to, tablets, capsules, gelcaps, and caplets that are adapted for controlled-release.
  • Extended release of neuregulin provides continuous therapeutic level of neuregulin over a period of time. For example neuregulin may be released over a period of 1 hour, 2 hours, 4 hours, 8 hours, 10 hours, 12 hours, 14 hours, 16 hours, 20 hours, 24 hours or longer. Alternatively, neuregulin may be released over a period of 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days or longer. Alternatively, neuregulin may be released over a period of 1 week, 2 weeks, 3 weeks, 4 weeks or longer. Alternatively, neuregulin may be released over a period of 1 month, 2 months, 4 months, 8 months, 12 months or longer. For example neuregulin may be released over a period of 1 years, 2 years, 3 years, 4 years or longer. Alternatively neuregulin is released over a period of between 1 hour and 2 week, between 2 hours and 2 week, between 4 hours to 24 hours, between 4 days and 10 days. The amount of time over which neuregulin is released may depend on various factors such as the extended release technology or technologies used. According to the invention, neuregulin is released over a period of between 4 hours and 12 hours per day by means of an osmotic pump or a syringe pump.
  • Extended release of neuregulin maintains neuregulin in the blood within a desirable range, particularly at a level which is at or above the minimum effective therapeutic level and is below the minimum toxic level over a period of time. The serum concentration of neuregulin in patients who received an extended release neuregulin composition can be compared with serum concentrations of patients receiving a non-extended release neuregulin composition (e.g., intravenous administration) at a time when the maximum blood level concentration occurs (Cmax). In a preferred embodiment, the patients receiving an extended release neuregulin composition have a lower maximum serum concentration (Cmax) of neuregulin than the patients receiving a non-extended neuregulin composition. Preferably, the patients receiving an extended release neuregulin composition have a Cmax less than about 90%, 80%, 70% or 60% of the Cmax in patients receiving a non-extended release neuregulin composition. More preferably, the patients receiving an extended release neuregulin composition have a Cmax less than about 50%, 40% or 30% of the Cmax in patients receiving a non-extended release neuregulin composition. Most preferably, the patients receiving an extended release neuregulin composition have a Cmax less than about 20%, 10% or less of the Cmax in patients receiving a non-extended release neuregulin composition. Methods for measuring the concentration of neuregulin in the serum are known in the art For instance, cells expressing ErbB-2 and ErbB-3 receptors, such as SKBR-3 breast cancer cell line, can be used. 10, 5, 2.5, 1.25, 0.625, 0.312, 0.156, 0.078, 0.039, 0.019 and 0.0079ng of neuregulin is added to different tubes containing cells separately on ice, then radiolabeled neuregulin (50,000cpm) is added. The sample solution is mixed and left at 4°C overnight. Next morning, cells are pelleted and the supernatant is sucked away before the radioactivity is counted. A standard curve is drawn by radioactivity versus unlabeled neuregulin amount When measuring the concentration of neuregulin in the serum, certain amount of serum is added to tube containing cells on ice, radiolabeled neuregulin (50,000cpm) is then added, and the sample solution is mixed and left at 4°C overnight. Next morning, cells are pelleted and the supernatant is sucked away before the radioactivity is counted. The radioactivity is counted and the amount of neuregulin in the serum can be calculated according to the standard curve.
  • Various extended release profiles can be provided in accordance with the present invention. "Extended release profile" means a release profile in which less than 50% of the total release of neuregulin that occurs over the course of implantation/insertion or other method of administering neuregulin in the body occurs within the first 24 hours of administration. In a preferred embodiment of the present invention, the extended release profile is selected from the group consisting of: (a) the 50% release point occurring at a time that is between 24 and 48 hours after implantation/insertion or other method of administration, (b) the 50% release point occurring at a time that is between 48 and 96 hours after implantation/insertion or other method of administration, (c) the 50% release point occurring at a time that is between 96 and 168 hours (1 week) after implantation/insertion or other method of administration, (d) the 50% release point occurring at a time that is between 1 and 2 weeks after implantation/insertion or other method of administration, (e) the 50% release point occurring at a time that is between 2 and 4 weeks after implantation/insertion or other method of administration, (f) the 50% release point occurring at a time that is between 4 and 8 weeks after implantation/insertion or other method of administration, (g) the 50% release point occurring at a time that is between 8 and 16 weeks after implantation/insertion or other method of administration, (h) the 50% release point occurring at a time that is between 16 and 52 weeks (1 year) after implantation/insertion or other method of administration, and (i) the 50% release point occurring at a time that is between 52 and 104 weeks after implantation/insertion or other method of administration.
  • Additionally, use of the present invention can reduce the degree of fluctuation ("DFL") of an agent's plasma concentration. DFL is a measurement of how much plasma levels of a drug vary over the course of a dosing interval. The closer the DFL is to zero (0), the less variance there is over the course of a dosing period. Thus a reduced DFL signifies that the difference in peak and trough plasma levels has been reduced. Preferably, the patients receiving an extended release composition have a DFL approximately 90%, 80%, 70% or 60% of the DFL in patients receiving a non-extended release composition. More preferably, the patients receiving an extended release composition have a DFL approximately 50%, 40%, or 30% of the DFL in patients receiving a non-extended release composition. Most preferably, the patients receiving an extended release neuregulin composition have a DFL approximately 20%, 10% or less of the DFL in patients receiving a non-extended release neuregulin composition.
  • Generally, the size and frequency of dosing is determined by the pharmacodynamic and pharmacokinetic properties of the active agent. The slower the rate of absorption, the less the blood concentrations fluctuate within a dosing interval. This' enables higher doses to be given less frequently. However, many active agents that are readily soluble in the body are usually absorbed rapidly and provide a sudden burst of available drug. An example is hypotension patients taking rapid-release nifedipine products. The use of an extended-release product avoids the high initial blood concentrations which cause the sudden reduction in blood pressure and other significant haemodynamic changes such as reflex tachycardia.
  • Additionally, some active agents are targeted and removed or destroyed by the body, e.g., immune system, proteases. Drugs with short half-lives for this and other reasons often need to be given the active agent at frequent intervals to maintain blood concentrations within the therapeutic range. There is an inverse correlation between the frequency of dosing and patient compliance. For such agents with relatively short half-lives, the use of extended-release products may maintain therapeutic concentrations over prolonged periods. Thus, a reduction in the number of daily doses offered by extended-release products has the potential to improve compliance. Although specific extended release technologies are disclosed herein, the invention is more general than any specific extended release technology. This includes the discovery that extended release of NRG at low doses unexpectedly improves the function of infarct heart. Further, there are numerous extended release drug delivery technologies currently known in the art. Several are generally discussed below as preferred extended release technologies, but they are offered solely for purposes of illustration and not limitation. Many other related and unrelated technologies are well known in the art and may be employed in the practice of the invention disclosed herein. Additionally, combinations of the extended release technologies discussed herein and/or other extended release technologies known in the art may be employed in the practice of this invention. For example, many companies with specific expertise in extended release drug delivery technologies - e.g., Alza Corp., Durect Corp., Gilead Sciences, Baxter Pharmaceuticals, Brookwood Pharmaceuticals and OctoPlus - offer products and services that can be employed in the practice of this invention. Additionally, a search of patents, published patent applications and related publications will provide those skilled in the art reading this disclosure with significant possible extended release technologies. Thus, one skilled in the art will be able to select the desired extended release technology or technologies for use in the practice of this invention.
  • C.1. Osmotic Pumps
  • In one embodiment of the present invention, the extended release of NRG into the blood comprises the use of an osmotic pump. Osmotic devices have demonstrated utility in delivering beneficial active agents to a target area in a controlled manner over prolonged periods of time. Known devices include tablets, pills, capsules and implantable devices. Tablets and pills can be taken orally, whereas other pumps are implanted subcutaneously or intraperitoneally, or attached to a catheter for intravenous, intracerebral or intra-arterial infusion.
  • Generally, in an osmotic pump system, a core is encased by a semipermeable membrane having at least one orifice. The semipermeable membrane is permeable to water, but impermeable to the active agent. When the system is exposed to body fluids, water penetrates through the semipermeable membrane into the core containing osmotic excipients and the active agent Osmotic pressure increases within the core and the agent is displaced through the orifice at a controlled, predetermined rate.
  • In many osmotic pumps, the core contains more than one internal compartment. For example, a first compartment may contain the active agent. A second compartment contains an osmotic agent and/or "driving member." See, e.g., U.S. Pat. No. 5,573,776 , the contents of which are incorporated herein by reference. This compartment may have a high osmolality, which causes water to flux into the pump through the semipermeable membrane. The influx of water compresses the first compartment. This can be accomplished, for example, by using a polymer in the second compartment, which swells on contact with the fluid. Accordingly, the agent is displaced at a predetermined rate.
  • In another embodiments, the osmotic pump may comprise more than one active agent-containing compartment, with each compartment containing the same agent or a different agent The concentrations of the agent in each compartment, as well as the rate of release, may also be the same or different.
  • The rate of delivery is generally controlled by the water permeability of the semipermeable membrane. Thus, the delivery profile of the pump is independent of the agent dispensed, and the molecular weight of an agent, or its physical and chemical properties, generally have no bearing on its rate of delivery. Further discussion regarding the principle of operation, the design criteria, and the delivery rate for osmotic pumps is provided in Theeuwes and Yum, Annals of Biomedical Engineering, Vol. 4, No. 4 (1976) and Urquhart et. al., Ann. Rev. Pharmacol. Toxicol. 24:199-236 (1984).
  • Osmotic pumps are well know in the art and readily available to one of ordinary skill in the art from companies experienced in providing osmotic pumps for extended release drug delivery. For example, ALZA's DUROS® technology is an implantable, nonbiodegradable, osmotically driven system that enables delivery of small drugs, peptides, proteins, DNA and other bioactive macromolecules for up to one year; ALZA's OROS® technology embodies tablets that employ osmosis to provide precise, controlled drug delivery for up to 24 hours; Osmotica Pharmaceutical's Osmodex® system includes a tablet, which may have more than one layer of the drug(s) with the same or different release profiles; Shire Laboratories' EnSoTrol® system solubilizes drugs within the core and delivers the solubilized drug through a laser-drilled hole by osmosis; and Alzet ® Osmotic pumps are miniature, implantable pumps used for research in mice, rats and other laboratory animals.
  • A search of patents, published patent applications and related publications will also provide those skilled in the art reading this disclosure with significant possible osmotic pump technologies. For example, U.S. Pat. Nos. 6,890,918 ; 6,838,093 ; 6,814,979 ; 6,713,086 ; 6,534,090 ; 6,514,532 ; 6,361,796 ; 6,352,721 ; 6,294,201 ; 6,284,276 ; 6,110,498 ; 5,573,776 ; 4,200,0984 ; and 4,088,864 , describe osmotic pumps and methods for their manufacture. One skilled in the art, considering both the disclosure of this invention and the disclosures of these other patents could produce an osmotic pump for the extended release of NRG.
  • Typical materials for the semipermeable membrane include semipermeable polymers known to the art as osmosis and reverse osmosis membranes, such as cellulose acylate, cellulose diacylate, cellulose triacylate, cellulose acetate, cellulose diacetate, cellulose triacetate, agar acetate, amylase triacetate, beta glucan acetate, acetaldehyde dimethyl acetate, cellulose acetate ethyl carbamate, polyamides, plyurethanes, sulfonated polystyrenes, cellulose acetate pphthalate, cellulose acetate methyl carbamate, cellulose acetate succinate, cellulose acetate dimethyl aminoacetate, cellulose acetate ethyl carbamate, cellulose acetate chloracetate, cellulose dipalmitate, cellulose dioctanoate, cellulose dicaprylate, cellulose dipentanlate, cellulose acetate valerate, cellulose acetate succinate, cellulose propionate, succinate, methyl cellulose, cellulose acetate p-toluene sulfonate, cellulose acetate butyrate, cross-linked selectively semipermeable polymers formed by the coprecipitation of a polyanion and a polycation, semipermeable polymers, lightly cross-linked polystyrene derivatives, cross-linked poly(sodium styrene sulfonate), poly(vinylbenzyltrimethyl ammonium chloride), cellulose acetate having a degree of substitution up to 1 and an acetyl content up to 50%, cellulose diacetate having a degree of substitution of 1 to 2 and an acetyl content of 21 to 35%, cellulose triacetate having a degree of substitution of 2 to 3 and an acetyl content of 35 to 44.8%, as disclosed in U.S. Pat. No. 6,713,086 .
  • The osmotic agent(s) present in the pump may comprise any osmotically effective compound(s) that exhibit an osmotic pressure gradient across the semipermeable wall against the exterior fluid. Effective agents include, without limitation, magnesium sulfate, calcium sulfate, magnesium chloride, sodium chloride, lithium chloride, potassium sulfate, sodium carbonate, sodium sulfite, lithium sulfate, potassium chloride, sodium sulfate, d-mannitol, urea, sorbitol, inositol, raffinose, sucrose, flucose, hydrophilic polymers such as cellulose polymers, mixtures thereof, and the like, as disclosed in U.S. Pat No. 6,713,086 .
  • The "driving member" is typically a hydrophilic polymer which interacts with biological fluids and swells or expands. The polymer exhibits the ability to swell in water and retain a significant portion of the imbibed water within the polymer structure. The polymers swell or expand to a very high degree, usually exhibiting a 2 to 50 fold volume increase. The polymers can be non-crosslinked or crosslinked. Hydrophilic polymers suitable for the present purpose are well known in the art.
  • The orifice may comprise any means and methods suitable for releasing the active agent from the system. The osmotic pump may include one or more apertures or orifices which have been bored through the semipermeable membrane by mechanical procedures known in the art, including, but not limited to, the use of lasers as disclosed in U.S. Pat. No 4,088,864 . Alternatively, it may be formed by incorporating an erodible element, such as a gelatin plug, in the semipermeable membrane.
  • Although specific embodiments of osmotic pumps are discussed above, the invention is more general than any specific extended release technology. This includes the discovery that extended release of NRG improves the function of infarct heart and reduces the interior diameter of the left ventricle. There are numerous variations and different types of osmotic pumps currently known in the art and may be employed in the practice of the invention disclosed herein.
  • C.2. Poly(ethylene glycol) Coupling
  • Disclosed is that the extended release of NRG into the blood comprises the coupling of the active agent to a polymer, such as Poly(ethylene glycol) (hereinafter referred to as "PEG"). Coupling PEG to biologically active agents has demonstrated utility in delivering active agents to a target area in a controlled manner over prolonged periods of time. Particularly, modification of proteins with PEG has been extensively used within the biotechnology industry to reduce the antigenicity of therapeutically active agents and to extend their in vivo availability. For example, coupling PEG to bovine adenosine deaminase using cyanuric chloride results in a loss of immunogenicity. Similarly, the PEG adduct of both human growth hormone and E. coli L-asparaginase has been shown to have an extended circulatory half-life.
  • Coupling PEG to an active agent or other molecules, e.g., outer surface of liposomes, can improve the efficacy and half-life of the active agent or other molecule, and also reduce its toxicity. Particularly, in an aqueous medium, the PEG molecule is hydrated and in rapid motion. This rapid motion causes the PEG to sweep out a large volume and prevents the approach and interference of other molecules, e.g., immune cells or proteases. Thus, when coupled to PEG, the PEG polymer chains can protect the attached molecule from immune response and other clearance mechanisms, sustaining availability of the active agent.
  • Generally, polyethylene glycol molecules are connected to the protein via a reactive group found on the protein. Commonly amino groups, such as those on lysine residues or at the N-terminus, are used for attachment. U.S. Pat. Nos. 5,824,784 and 4,002,531 disclose such methods for attaching PEG to an enzyme by reductive alkylation. Lysine residues may be strategically substituted for other amino acids or inserted into a polypeptide sequence to provide additional points of attachment as disclosed in U.S. Pat: No. 4,904,584 . Additional methods are known in the art for attaching branched or "multi-armed" PEG-derivatives to proteins as disclosed in U.S. Pat. No. 5,932,462 . There are many other methods of attachment known in the art for attaching polymers to cysteine residues, carboxy groups, carbohydrates and other moieties. For example, U.S. Pat. No. 5,900,461 discloses derivatives of PEG and other polymers having one more active sulfone moieties that are highly selective for coupling with thiol moieties instead of amino moieties on molecules.
  • PEGs can also be used to link macromolecules to a targeting ligand or moiety, which directs the macromolecules to particular areas of interest. U.S. Pat. No. 6,436,386 discloses active agent-polymer conjugates attached to a hydroxyapatite-targeting moiety for delivery of the active agent, such as bone growth factors, to hydroxyapatite surfaces, such as bone.
  • A wide variety of PEG derivatives are both available and suitable for use in the preparation of PEG-conjugates. For example, NOF Corp.'s SUNBRIGHT® Series provides numerous PEG derivatives, including methoxypolyethylene glycols and activated PEG derivatives, such as methoxy-PEG amines, maleimides and carboxylic acids, for coupling by various methods to drugs; enzymes, phospholipids and other biomaterials and Nektar Therapeutics' Advanced PEGylation also offers diverse PEG-coupling technologies to improve the safety and efficacy of therapeutics.
  • A search of patents, published patent applications and related publications will also provide those skilled in the art reading this disclosure with significant possible PEG-coupling technologies and PEG-derivatives. For example, U.S. Pat. Nos. 6,436,386 ; 5,932,462 ; 5,900,461 ; 5,824,784 ; 4,904,584 and 4,002,531 , describe such technologies and derivatives, and methods for their manufacture. Thus, one skilled in the art, considering both the disclosure of this invention and the disclosures of these other patents could couple PEG, a PEG-derivative or some other polymer to NRG for its extended release.
  • PEG is a well known polymer having the properties of solubility in water and in many organic solvents, lack of toxicity, lack of immunogenecity, and also clear, colorless, odorless and stable. One use of PEG is to covalently attach the polymer to insoluble molecules to make the resulting PEG-molecule conjugate soluble. For these reasons and others, PEG has been selected as the preferred polymer for attachment, but it has been employed solely for purposes of illustration and not limitation. Similar products may be obtained with other water soluble polymers, including without limitation, poly(vinyl alcohol), other poly(alkylene oxides) such as poly(propylene glycol) and the like, poly(oxyethylated polyols) such as poly(oxyethylated glycerol) and the like, carboxymethylcellulose, dextran, polyvinyl alcohol, polyvinyl pyrrolidone, poly-1,3-dioxolane, poly-1,3,6-trioxane, ethylene/maleic anhydride, and polyaminoacids. One skilled in the art will be able to select the desired polymer based on the desired dosage, circulation time, resistance to proteolysis, and other considerations.
  • C. 3. Liposome Packaging
  • Disclosed in that the extended release of NRG into the blood comprises packaging NRG in a liposome, which has demonstrated utility in delivering beneficial active agents in a controlled manner over prolonged periods of time. Liposomes are completely closed bilayer membranes containing an entrapped aqueous volume. Liposomes may be unilamellar vesicles possessing a single membrane bilayer or multilamellar vesicles with multiple membrane bilayers, each separated from the next by an aqueous layer. The structure of the resulting membrane bilayer is such that the hydrophobic (non-polar) tails of the lipid orient toward the center of the bilayer while the hydrophilic (polar) heads orient towards the aqueous phase.
  • Generally, in a liposome-drug delivery system, the active agent is entrapped in the liposome and then administered to the patient to be treated. However, if the active agent is lipophilic, it may associate with the lipid bilayer.
  • The immune system may recognize conventional liposomes as foreign bodies and destroy them before significant amounts of the active agent reaches the intended disease site. Thus, in one embodiment, the liposome may be coated with a flexible water soluble polymer that avoids uptake by the organs of the mononuclear phagocyte system, primarily the liver and spleen. Suitable hydrophilic polymers for surrounding the liposomes include, without limitation, PEG, polyvinylpyrrolidone, polyvinylmethylether, polymethyloxazoline, polyethyloxazoline, polyhydroxypropyloxazoline, polyhydroxypropylmethacrylamide, polymethacrylamide, polydimethylacrylamide, polyhydroxypropylmethacrylate, polyhydroxethylacrylate, hydroxymethylcellulose hydroxyethylcellulose, polyethyleneglycol, polyaspartamide and hydrophilic peptide sequences as described in U.S. Pat. Nos. 6,316,024 ; 6,126,966 ; 6,056,973 ; 6,043,094 .
  • Liposomes may be comprised of any lipid or lipid combination known in the art. For example, the vesicle-forming lipids may be naturally-occurring or synthetic lipids, including phospholipids, such as phosphatidylcholine, phosphatidylethanolamine, phosphatidic acid, phosphatidylserine, phasphatidylglycerol, phosphatidylinositol, and sphingomyelin as disclosed in U.S. Patent Nos. 6,056,973 and 5,874,104 . The vesicle-forming lipids may also be glycolipids, cerebrosides, or cationic lipids, such as 1,2-dioleyloxy-3-(trimethylamino) propane (DOTAP); N-[1-(2,3,-ditetradecyloxy)propyl]-N,N-dimethyl-N-hydroxyethylammonium bromide (DMRIE); N-[1[(2,3,-dioleyloxy)propyl]-N,N-dimethyl-N-hydroxy ethylammonium bromide (DORIE); N-[1-(2,3-dioleyloxy)propyl]-N,N,N-trimethylammonium chloride (DOTMA); 3 [N-(N',N'-dimethylaminoethane) carbamoly] cholesterol (DC-Chol); or dimethyldioctadecylammonium (DDAB) also as disclosed in U.S. Patent No. 6,056,973 . Cholesterol may also be present in the proper range to impart stability to the vesicle as disclosed in U.S. Pat. Nos. 5,916,588 and 5,874,104 .
  • The liposome may be targeted to specific sites within the body of a mammal by the attachment of a targeting ligand or moiety. The targeting ligands are believed to be recognized by receptors or other compounds on the surface of target cells. Typical target ligands include antibodies or antibody fragments, cell-receptor ligands, lectins and the like. For further discussion see U.S. Pat. Nos. 6,316,024 and 6,294,191 .
  • Such targeting ligands can be attached to liposomes by any means known in the art for the covalent or noncovalent attachment of such ligands to lipsomes. For example, polymer coated liposomes have been modified to achieve site specific delivery of active agents, by attaching a targeting ligand to either the polar head group residues of liposomal lipid components or the free ends of the polymer chains forming the surface coat on the liposomes as described in U.S. Patent No. 6,316,024 and 6,043,094 . Such attachments may be accomplished by, for example, the coupling of proteins to liposomes through the use of a crosslinking agent having at least one maleimido group and an amine reductive function as described in U.S. Pat. No. 5,399,331 ; linking proteins to liposomes through the use of the glycoprotein streptavidin as described in U.S. Patent Nos. 4,885,172 ; 5,059,421 and 5,171,578 ; the coating of targeted liposomes with polysaccharides; or a vesicle forming lipid may derivatized with a hydrophilic polymer chain, which is end-functionalized for coupling antibodies through the use of a hydrazide or hydrazine group that is reactive toward aldehyde groups as described in U.S. Pat. No. 6,126,966 . The end functionalized group may also be 2-pyridyidithio-propionamide, for coupling an antibody or other molecule to the liposome through a disulfide linkage.
  • The liposomes can be manufactured by standard techniques known to those of skill in the art. For example, in one embodiment, as disclosed in U.S. Pat. No. 5,916,588 , a buffered solution of the active agent is prepared. Then a suitable lipid, such as hydrogenated soy phosphatidylcholine, and cholesterol, both in powdered form, are dissolved in chloroform or the like and dried by rotoevaporation. The lipid film thus formed is resupsended in diethyl ether or the like and placed in a flask, and sonicated in a water bath during addition of the buffered solution of the active agent. Once the ether has evaporated, sonication is discontinued and a stream of nitrogen is applied until residual ether is removed. Other standard manufacturing procedures are described in U.S. Pat. Nos. 6,352,716 ; 6,294,191 ; 6,126,966 ; 6,056,973 ; 5,965,156 ; and 5,874,104 . The liposome can be produced by any method generally accepted in the art for making liposomes, including, without limitation, the methods of the above-cited documents.
  • Liposomes are also well known in the art and readily available from companies experienced in providing liposomes for extended release drug delivery. For example, ALZA's (formerly Sequus Pharmaceuticals') STEALTH® liposomal technology for intravenous drug delivery uses a polyethylene glycol coating on liposomes to evade recognition by the immune system; Gilead Sciences (formerly Nexstar's) liposomal technology was incorporated into AmBisome®, and FDA approved treatment for fungal infections; and NOF Corp. offers a wide variety of GMP-grade phospholipids, phospholipids derivatives, and PEG-phospholipids under the tradenames COATSOME® and SUNBRIGHT®.
  • A search of patents, published patent applications and related publications will also provide those skilled in the art reading this disclosure with significant possible liposomal technologies. U.S. Pat. Nos. 6,759,057 ; 6,406,713 ; 6,352,716 ; 6,316,024 ; 6,294,191 ; 6,126,966 ; 6,056,973 ; 6,043,094 ; 5,965,156 ; 5,916,588 ; 5,874,104 ; 5,215,680 ; and 4,684,479 , describe liposomes and lipid-coated microbubbles, and methods for their manufactured.
  • C. 4. Microsphere Packaging
  • Disclosed is that the extended release of NRG into the blood comprises packaging NRG in a microsphere. Microspheres have demonstrated utility in delivering beneficial active agents to a target area in a controlled manner over prolonged periods of time. Microspheres are generally biodegradable and can be used for subcutaneous, intramuscular and intravenous administration.
  • Generally, each microsphere is composed of an active agent and polymer molecules. As disclosed in U.S. Pat No. 6,268,053 , the active agent may be centrally located within a membrane formed by the polymer molecules, or, alternatively dispersed throughout the microsphere because the internal structure comprises a matrix of the active agent and a polymer excipient. Typically, the outer surface of the microsphere is permeable to water, which allows aqueous fluids to enter the microsphere, as well as solubilized active agent and polymer to exit the microsphere.
  • The polymer membrane may comprise crosslinked polymers as disclosed in U.S. Patent No. 6,395,302 . When the pore sizes of the crosslinked polymer are equal or smaller than the hydrodynamic diameter of the active agent, the active agent is essentially released when the polymer is degraded. On the other hand, if the pore size of the crosslinked polymers are larger than the size of the active agent, the active agent is at least partially released by diffusion.
  • Additional methods for making microsphere membranes are known and used in the art. Typical materials for the outer membrane include the following categories of polymers: (1) carbohydrate-based polymers, such as methylcellulose, carboxymethyl cellulose-based polymers, dextran, polydextrose, chitins, chitosan, and starch (including hetastarch), and derivatives thereof; (2) polyaliphatic alcohols such as polyethylene oxide and derivatives thereof including polyethylene glycol (PEG), PEG-acrylates, polyethyleneimine, polyvinyl acetate, and derivatives thereof; (3) poly(vinyl) polymers such as poly(vinyl) alcohol, poly(vinyl)pyrrolidone, poly(vinyl)phosphate, poly(vinyl)phosphonic acid, and derivatives thereof; (4) polyacrylic acids and derivatives thereof; (5) polyorganic acids, such as polymaleic acid, and derivatives thereof; (6) polyamino acids, such as polylysine, and poly-imino acids, such as polyimino tyrosine, and derivatives thereof; (7) co-polymers and block co-polymers, such as poloxamer 407 or Pluronic L-101™ polymer, and derivatives thereof; (8) tert-polymers and derivatives thereof; (9) polyethers, such as poly(tetramethylene ether glycol), and derivatives thereof; (10) naturally occurring polymers, such as zein, chitosan and pullulan, and derivatives thereof; (11) polyimids, such as poly n-tris(hydroxymethyl) methylmethacrylate, and derivatives thereof; (12) surfactants, such as polyoxyethylene sorbitan, and derivatives thereof; (13) polyesters such poly(ethylene glycol) (n)monomethyl ether mono(succinimidyl succinate)ester, and derivatives thereof; (14) branched and cyclo-polymers, such as branched PEG and cyclodextrins, and derivatives thereof; and (15) polyaldehydes, such as poly(perfluoropropylene oxide-b-perfluoroformaldehyde), and derivatives thereof as disclosed in U.S. Pat. No. 6,268,053 . Other typical polymers known to those of ordinary skill in the art include poly(lactide-co-glycolide, polylactide homopolymer; polyglycolide homopolymer; polycaprolactone; polyhydroxybutyrate-polyhydroxyvalerate copolymer; poly(lactide-co-caprolactone); polyesteramides; polyorthoesters; poly 13-hydroxybutyric acid; and polyanhydrides as disclosed in U.S. Pat. No. 6,517,859 .
  • In one embodiment, the microspheres are attached to or coated with additional molecules. Such molecules can facilitate targeting, enhance receptor mediation, and provide escape from endocytosis or destruction. Typical molecules include phospholipids, receptors, antibodies, hormones and polysaccharides. Additionally, one or more cleavable molecules may be attached to the outer surface of microspheres to target it to a predetermined site. Then, under appropriate biological conditions, the molecule is cleaved causing release of the microsphere from the target.
  • The microspheres are manufactured by standard techniques. For example, in one embodiment, volume exclusion is performed by mixing the active agent in solution with a polymer or mixture of polymers in solution in the presence of an energy source for a sufficient amount of time to form particles as disclosed in U.S. Pat. No. 6,268,053 . The pH of the solution is adjusted to a pH near the isoelectric point (pI) of the macromolecule. Next, the solution is exposed to an energy source, such as heat, radiation, or ionization, alone or in combination with sonication, vortexing, mixing or stirring, to form microparticles. The resulting microparticles are then separated from any unincorporated components present in the solution by physical separation methods well known to those skilled in the art and may then be washed. Other standard manufacturing procedures are described in U.S. Pat. Nos. 6,669,961 ; 6,517,859 ; 6,458,387 ; 6,395,302 ; 6,303,148 ; 6,268,053 ; 6,090,925 ; 6,024,983 ; 5,942,252 ; 5,981,719 ; 5,578,709 ; 5,554,730 ; 5,407,609 ; 4,897,268 ; and 4,542,025 .
  • Microspheres are well known and readily available to one of ordinary skill in the art from companies experienced in providing such technologies for extended release drug delivery. For example, Epic Therapeutics, a subsidiary of Baxter Healthcare Corp., developed PROMAXX®, a protein-matrix drug delivery system that produces bioerodible protein microspheres in a totally water-based process; OctoPlus developed OctoDEX®, crosslinked dextran microspheres that release active ingredients based on bulk degradation of matrix rather than based on surface erosion; and Brookwood Pharmaceuticals advertises the availability of its microparticle technologies for drug delivery.
  • A search of patents, published patent applications and related publications will also provide those skilled in the art reading this disclosure with significant possible microsphere technologies. For example, U.S. Pat. Nos. 6,669,961 ; 6,517,859 ; 6,458,387 ; 6,395,302 ; 6,303,148 ; 6,268,053 ; 6,090,925 ; 6,024,983 ; 5,942,252 ; 5,981,719 ; 5,578,709 ; 5,554,730 ; 5,407,609 ; 4,897,268 ; and 4,542,025 describe microspheres and methods for their manufacture.
  • D. Dosage and Frequency of Administration
  • The amount of neuregulin used in the present invention will vary with the nature and severity of the disease or condition, and the route by which the active ingredient is administered. The frequency and dosage will also vary according to factors specific for each patient depending on the specific therapy (e.g., therapeutic or prophylactic agents). administered, the severity of the disorder, disease, or condition, the route of administration, as well as age, body, weight, response, and the past medical history of the patient. Effective doses may be extrapolated from dose-response curves derived from in vitro or animal model test systems.
  • Exemplary doses of neuregulin include milligram or microgram amounts of neuregulin per kilogram of subject or sample weight (e.g. about 1 microgram per kilogram to about 500 milligrams per kilogram, about 100 micrograms per kilogram to about 5 milligrams per kilogram, or about 1 microgram per kilogram to about 50 micrograms per kilogram). For extened release of neuregulin used in the invention, the dosage administered to a patient is typically 0.001 mg/kg to 15 mg/kg of the patient's body weight, based on weight of the active peptide. Preferably, the dosage administered to a patient is between 0.001 mg/kg and 15 mg/kg, 0.005 mg/kg and 10 mg/kg, 0.01 mg/kg and 5 mg/kg, 0.001 mg/kg and 4 mg/kg, 0.005 mg/kg and 3 mg/kg, 0.01 mg/kg and 2 mg/kg, 0.001 mg/kg and 1 mg/kg, 0.005 mg/kg and 0.5 mg/kg, 0.010 mg/kg and 0.2 mg/kg, 0.005 mg/kg and 0.050 mg/kg of the patient's body weight.
  • Exemplary doses of neuregulin also include unit (U) or unit amounts of neuregulin per kilogram of subject or sample weight (e.g., about 1 U per kilogram to about 5000 U per kilogram, about 10 U micrograms per kilogram to about 1000 per kilogram, or about 100 U per kilogram to about 500 U per kilogram). For extened release of neuregulin used in the invention, the dosage administered to a patient is typically 10 U/kg to 1000 U/kg of the patient's body weight, based on weight of the active peptide. Preferably, the dosage administered to a patient is between 1 U/kg and 10,000U/kg, 1 U/kg and 5000 U/kg, 10 U/kg and 5000 U/kg, 10 U/kg and 1000 U/kg, 50 U/kg and 2000 U/kg, 50 U/kg and 1000/kg, 50 U/kg and 500 U/kg, 100 U/kg and 1000 U/kg, 100 U/kg and 500 U/kg, 100 U/kg and 200 U/kg, of the patient's body weight.
  • In general, the recommended daily dose range of neuregulin in the methods of the invention for the conditions described herein lie within the range of from about 0.001 mg to about 1000 mg per day. Specifically, a total daily dose range should be between 0.001 mg per day and 15 mg per day, 0.005 mg per day and 10 mg per day, 0.01 mg per day and 5 mg per day, 0.001 mg per day and 4 mg per day, 0.005 mg per day and 3 mg per day; 0.01 mg per day and 2 mg per day, 0.001 mg per day and 1 mg per day, 0.005 mg per day and 0.5 mg per day, 0.010 mg per day and 0.2 mg per day. In managing the patient, the therapy can be initiated at a lower dose, perhaps about 0.1 µg to about 1 µg, and increased if necessary up to about 20µg mg to about 1000 µg per day as either a single dose or divided doses, depending on the patient's global response. It may be necessary to use dosages of the active ingredient outside the ranges disclosed herein in some cases, as will be apparent to those of ordinary skill in the art. Furthermore, it is noted that the clinician or treating physician will know how and when to interrupt, adjust, or terminate therapy in conjunction with individual patient response. In certain embodiments, neuregulin is administered in an amount of about 1 U/day to about 10,000 U/day. In some embodiments, it is administered in an amount of about 1 U/day to about 5000 U/day. In some embodiments, it is administered in an amount of about 10U/day to about 2000 U/day. In some embodiments, it is administered in an amount of about 10 U/day to about 1000 U/day. In some embodiments, it is administered in an amount of about 100 U/day to about 200 U/day.
  • Neuregulin can also be administered in a dosing schedule or "therapeutic cycle." Daily dosage of neuregulin in the therapeutic cycle is described in detail above. The therapeutic cycle can last 2 days, 5 days, 7 days, 10 days, two weeks, three weeks, four weeks, five weeks, or six weeks.
  • In certaine embodiments, neuregulin is administered daily for each day of the therapeutic cycle. In certain embodiments, neuregulin is administered consecutively for three, four, five, six, seven, eight, nine, ten, eleven or twelve days in a therapeutic cycle.
  • In certain embodiments, in a therapeutic cycle neuregulin is administered on day 1 of the cycle and the cycle concludes with one or more days of no neuregulin administration In some embodiments, neuregulin is administered daily for 3, 5, 7, or 10 days followed by a resting period in a therapeutic cycle.
  • E. Combinational Therapy
  • In one embodiment, the present invention is useful in preventing heart failure in patients being treated with a drug that causes cardiac hypertrophy or heart failure, e.g., fludrocortisone acetate or herceptin. NRG may be administered prior to, simultaneously with, or subsequent to a drug which causes such cardiac diseases.
  • In another embodiment of the invention, NRG is administered in combination with an effective amount of a compound that acts to suppress a different hypertrophy induction pathway than NRG. In an alternative embodiment, NRG is administered with such hypertrophy suppressors and/or additional components, without limitation, a cardiotrophic inhibitor such as a Ct-1 (cardiotrophin-1) antagonist, an ACE inhibitor, such as captopril (Capoten®), and/or human growth hormone and/or IGF-I (Insulin like growth factor I) in the case of congestive heart failure, or with another anti-hypertrophic, myocardiotrophic factor, anti-arrhythmic, or inotropic factor in the case of other types of heart failure or cardiac disorder.
  • In another embodiment of the invention, NRG is administered in combination with current therapeutic approaches for treatment of heart failure, including, without limitation, ACE inhibitors and other vasodilators, diuretics, digitalis preparations, beta blockers, blood thinners, angiotensin II receptor blockers, calcium channel blockers or potassium.
  • ACE inhibitors, which prevent the conversion of angiotensin I to angiotensin II, are vasodilators that cause the blood vessels to expand, lowering the blood pressure and reducing the heart's workload. Vasodilators suitable for use in embodiments of the present invention include, without limitation, the following drugs: quinapril (Accupril®), ramipril (Altace®), captopril (Capoten®), benazepril (Lotensin®), fosinopril (Monopril®), lisinopril (Prinivil® or Zestril®), enalapril (Vasotec®), moexipril (Univasc®), trandolapril, and perindopril. Additional vasodilators useful in the present invention, include, without limitation, isosorbide dinitrate (Isordil®), nesiritide (Natrecor®), hydralazine (Apresoline®), nitrates and minoxidil.
  • Diuretics cause the kidneys to remove sodium and water from the blood stream, reducing the heart's workload, and include, without limitation, the following drugs: hydrochlorothiazide (HydroDIURIL®), chlorothiazide (Diuril®), furosemide (Lasix®), bumetanide (Bumex®), spironolactone (Aldactone®), triamterene (Dyrenium®); metolazone (Zaroxolyn®), torsemide, indapamide, polythiazide, amiloride, and combination agents (Dyazide®).
  • Digitalis preparations increase the force of the heart's contractions and include, without limitation, digoxin (Lanoxin®) and digitoxin.
  • Beta blockers reduce the heart's tendency to beat faster and include, without limitation, the following drugs: carvedilol (Coreg®) metoprolol (Lopressor® or Toprol XL®, atenolol, bisoprolol, labetalol, propranolol, sotalol, pindolol, penbutolol, acebutolol, timolol, nadolol, and betaxolol.
  • Blood thinners for use in embodiments of the present invention, include, without limitation, warfarin (Coumadin®) and heparin.
  • Embodiments of the present invention may also use angiotensin II receptor blockers, which, rather than lowering the levels of angiotensin II (as ACE inhibitors do), prevents angiotensin II from effecting the heart and blood vessels. Angiotensin II receptor blockers suitable for use in the present invention, include, without limitation, iosartan (Cozaar®); valsartan (Diovan®), irbesartan (Avapro®), candesartan, eprosartan, telmisartan, and olmesartan.
  • Calcium channel blockers are generally used to treat high blood pressure often associated with heart failure. Calcium channel blockers suitable for use in the present invention include, without limitation, amlodipine (Norvasc®).
  • In alternative embodiments of the present invention, extended release of NRG can also be combined with the administration of drug therapies for the treatment of heart diseases such as hypertension. For example, NRG can be administered with endothelin receptor antagonists, such as antibodies to the endothelin receptor, and peptides or other such small molecule antagonists; 3-adrenoreceptor antagonists such as carvedilol; x,-adrenoreceptor antagonists; anti-oxidants; compounds having multiple activities (e.g., 3-blocker/a-blocker/anti-oxidant); carvedilol-like compounds or combinations of compounds providing multiple functions found in carvedilol; growth hormone, etc.
  • Neuregulin agonists alone or in combination with other hypertrophy suppressor pathway agonists or with molecules that antagonize known hypertrophy induction pathways, are useful as drugs for in vivo treatment of mammals experiencing heart failure, so as to prevent or lessen heart failure effects.
  • Therapeutic formulations of agonist(s) for treating heart disorders are prepared for storage by mixing the agonist(s) having the desired degree of purity with optional physiologically acceptable carriers, excipients, or stabilizers ( Remington's Pharmaceutical Sciences, 16th edition, Oslo, A., Ed., 1980), in the form of lyophilized cake or aqueous solutions. Acceptable carriers, excipients, or stabilizers are non-toxic to recipients at the dosages and concentrations employed, and include buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid; low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or immunoglobins; hydrophilic polymers such as olyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, arginine or lysine; monosaccharides, disaccharides, and other carbohydrates including glucose, mannose, or dextrins; chelating agents such as EDTA; sugar alcohols such as mannitol or sorbitol; salt-forming counter ions such as sodium; and/or non-ionic surfactants such as Tween, Pluronics, or polyethylene glycol (PEG). The antagonist(s) are also suitably linked to one of a variety of nonproteinaceous polymers, e.g., polyethylene glycol, polypropylene glycol, or polyalkylenes, in the manner set forth in US Patent Nos. 4,640,835 ; 4,496,689 ; 4,301,144 ; 4,670,417 ; 4,791,192 or 4,179,337 . The amount of carrier used in a formulation may range from about 1 to 99%, preferably from about 80 to 99%, optimally between 90 and 99% by weight
  • The agonist(s) to be used for in vivo administration should be sterile. This is readily accomplished by methods known in the art, for example, by filtration through sterile filtration membranes, prior to or following lyophilization and reconstitution. The agonist(s) ordinarily will be stored in lyophilized form or in solution.
  • Therapeutic agonist compositions generally are placed into a container having a sterile access port, for example, an intravenous solution bag or yial having a stopper pierceable by a hypodermic injection needle. The agonist(s) administration is in a chronic fashion only, for example, one of the following routes: injection or infusion by intravenous, intraperitoneal, intracerebral, intramuscular, intraocular, intraarterial, or intralesional routes, orally or using sustained-release systems as noted above.
  • As discussed above, suitable examples of sustained-release preparations include. semipermeable matrices of solid hydrophobic polymers containing the protein, which matrices are in form of shaped articles, e.g., films, or microcapsules. Examples of sustained-release matrices include polyesters, hydrogels (e.g., poly(2-hydroxyethylmethacrylate) as described by Langer et al. (1981) J. Biomed. Mater. Res. 15: 167-277 and Langer (1982) Chem. Tech. 12: 98-105, or poly(vinyl alcohol)), polylactides ( US Patent No 3,773,919 , EP 58,481 ), copolymers of L-glutainic acid and gamma ethyl-L-. glutamate (Sidman et al. (1983) Biopolymers 22: 547-556), non-degradable ethylene-vinyl acetate (Langer et al. (1981) supra) degradable lactic acidglycolic acid copolymers such as the Lupron DepotTM (injectable microspheres composed of lactic acid-glycolic acid copolymer and leuprolide acetate), and poly-D-(-)-3-hydroxybutyric acid ( EP 133,988 ).
  • The agonist(s) also may be entrapped in microcapsules prepared, for example, by coacervation techniques or by interfacial polymerization (for example, hydroxymethylcellulose or gelatin-microcapsules and poly[methylmethacylate] microcapsules, respectively), in colloidal drug delivery systems (for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules), or in macroemulsions. Such techniques are disclosed in Remington's Pharmaceutical Sciences, supra.
  • While polymers such as ethylene-vinyl acetate and lactic acid-glycolic acid enable release of molecules for over 100 days, certain hydrogels release molecules for shorter time periods. When encapsulated molecules remain in the body for a long time, they may denature or aggregate as a result of exposure to moisture at 37°C, resulting in a loss of biological activity and possible changes in immunogenicity. Rational strategies can be devised for stabilization depending on the mechanism involved, e.g., using appropriate additives, and developing specific polymer matrix compositions.
  • Sustained-release agonist(s) compositions also include liposomally entrapped agonists(s). Liposomes containing agonists(s) are prepared by methods known in the art, for example, those disclosed in DE 3,218,121 ; Epstein et al. (1985) Proc. Natl. Acad. Sci. USA 82: 3688-3692; Hwang et al. (1980) Proc. Natl. Acad. Sci. USA 77:4030-4034: EP 52,322 ; EP 36,676 ; EP 88,046 ; EP 143,949 ; EP 142,641 ; Japanese patent application 83-118008 ; US Patent Nos. 4,485,045 and 4,544,545 ; and EP 102,324 . A specific example of suitable sustained-release formulation is in EP 647,449 .
  • In another embodiment of the present invention, NRG is combined with or administered in concert with other agents for treating congestive heart failure, including ACE inhibitors (as discussed above), CT-1 inhibitors, human growth hormone, and/or IGF-I. The effective amounts of such agents, if employed will be at the clinician's discretion. Dosage administration and adjustment are determined by methods known to those skilled in the art to achieve the best management of congestive heart failure and ideally takes into account use of diuretics or digitalis, and conditions such as hypotension and renal impairment. The dose will additionally depend on such factors as the type of drug used and the specific patient being treated. Typically the amount employed will be the same dose as that used if the drug were to be administered without agonist; however, lower doses may be employed depending on such factors as the presence of side-effects the condition being treated, the type of patient, and the type of agonists and drug, provided the total amount of agents provides an effective dose for the condition being treated.
  • F. Kits
  • The application relates to kits for carrying out the therapeutic regiments of the invention. Such kits comprise in one or more containers therapeutically effective amounts of NRG described herein, alone or in combination with other agents, in pharmaceutically acceptable form and in combination with an extended release technology as described herein. Instructions are optionally included for administration of the extended release NRG composition by a physician or by the patient.
  • G. Examples
  • As shown in the Examples, the invention resides in the discovery that extended release of NRG activates the AKT or ERK signaling pathway as effectively as NRG delivered by other methods, and improves the function of infarct heart much more than NRG delivered by other methods. However, NRG's interactions with ErbB receptors has been implicated in other diseases and disorders, e.g., diseases of the central and peripheral nervous system. Examples of other diseases and disorders, include, various cardiovascular diseases, cancer, neural system disease and/or muscle diseases, including muscular dystrophy (e.g., Duchenne, Limb-girdle) and multiple sclerosis, spinal injury, eye and ear diseases, diabetes, schizophrenia, and Alzheimer's.
  • The invention will be further illustrated by reference to the following non-limiting Examples. Efforts have been made to ensure accuracy with respect to numbers used but some experimental errors and deviations should be accounted for.
  • EXAMPLE 1 Phosphorylation of AKT and ERK in the left ventricle of normal rats after NRG is infused by different methods.
  • To compare the effect of NRG with various treating methods on the signal transduction inside the cardiac myocytes in the left ventricle, we infused NRG by intravenous (hereinafter referred to as "IV"), intramuscular (hereinafter referred to as "IM") and IV glucose tolerance test (hereinafter referred to as "IVGTT").
  • Wistar male rats (Shanghai Animal Center of Chinese Academy of Science), which weighed 180 ± 20 grams, were numbered, weighed, and divided into groups. Each group contained three rats. One group received IV injection of 4 ml/kg (volume/body weight) of vehicle (10mM Na2HPO4-NaH2PO4, 150mM NaCl, 0.2% human serum albumin (HSA), 5% mannitol, pH 6.0) as a control. Four other groups of rats received IM injection of 4 ml/kg (volume/body weight) of NRG (37.3U/ml recombinant human NRG fragment (from the 177th to 237th amino acid sequence of human NRG1β2 produced by Zensun Science & Technology - batch number 200503002)) dissolved in vehicle (as described above). Another four groups of rats received IV injection of 4 ml/kg (volume/body weight) of NRG (as described above). Another five groups of rats received 20µl/min of glucose tolerance test infusion of NRG (as described above) by IV injection (IVGTT) for two hours. Thus, the total amount of NRG administered to each rat (except for the vehicle group) was 149.3U/kg of body weight.
  • Rats were killed separately at 20min, 1hr, 2hr, 4hr and 6hr. The left ventricles of each group of rats were cut into pieces in cold lysis buffer (50mM Tris pH 7.4, 5mM EDTA, 150mM NaCl, 1% Triton X-100, 2mM Na2VO4, 50mM NaF, 2mM PMSF, protease inhibitor cocktail (no EDTA, Roche)) after pooled, and washed with cold PBS. The ventricles were then homogenized in ice water and centrifuged (Kendro Biofuge) at 12,000 rpm for 5 min at 4°C in 1.5ml Eppendorf tubes. The supernatant was collected and spun one more time, then stored at -80°C. The samples were thawed and spun again before use. The protein concentration of each sample was determined by BCA protein assay (Pierce BCA protein assay kit). A certain amount of each sample was mixed with 2X sample buffer (0.125M Tris ph 6.8, 20% glycerol, 4% SDS, 0.2M DTT, 0.012% bromophenol blue) and boiled for electrophoresis before transfer to PVDF membrane (Millipore). The phosphorylation of AKT and ERK, as well as the amount of AKT and ERK in each sample was detected with antibodies (ERK antibody and phosphorylated ERK antibody (Santa Cruz Biotechnology); AKT antibody and phosphorylated AKT antibody (Cell Signaling)).
  • The time course of phosphorylation of AKT and ERK in the left ventricle of normal rats when NRG was infused by each of these different methods is shown in Fig 1. Compared to the vehicle, NRG infused by IM, IV and IVGTT all activated sustained phosphorylation of ERK. AKT phosphorylation induced by each method peaked at 20 min and decreased at 1hr, but increased again at 2 hr, where it maintained a high level from 4 hr to 6 hr. Thus, there is no obvious difference among the different methods of injecting NRG with respect to their ability to sustain phosphorylation of ERK and AKT. This indicates that NRG infused constantly is as effective as injection of NRG. Thus, IVGTT infusion is a potential method for treating poor cardiac conditions.
  • EXAMPLE 2 The function of left ventricle coronary artery ligated rat heart after neuregulin treatment by different methods
  • As osmotic pump is a way to deliver NRG constantly (as IVGTT), we examined whether NRG infused by osmotic pump was as effective as conventional IV injection in restoring the function of myocardial infarct (MI) heart.
  • A. Rat left ventricle coronary artery ligation and echocardiography
  • Wistar male rats (Shanghai Animal Center of Chinese Academy of Science), which weighed 200 ± 20g, were anesthetized by intraperitoneally injecting 100mg/kg (drug/body weight) of ketamine. The neck and chest were depilated and sanitized. An incision was made in the middle front neck to expose the tracheae. An 18G catheter overneedle was inserted into the tracheae between the 3rd and 5th cartilage of tracheae. After the needle was drawn out, a plastic cannula was pushed into the trachea 1-2 cm and fixed to connect the Rodent Ventilator (SAR-830/P ventilator -Inspiratory flow rate, 1ml/100g/breath; Respiratory rate, 60 breaths/min). Another incision was made on the left front chest. The skin was blunt dissected to expose the fourth and fifth rib, then the fourth rib was cut by elbowed mosquito forceps. The ventilator (as described above) was linked to the cannula and turned on, and the heart was exposed to check the status of lung and heart. The pericardium was rived off to identify the left atria and the pulmonary arterious cone after the heart was exteriorized through the incision. The left ventricle anterior descending coronary artery between them was ligated tight with 6/0 medical suture before the heart was replaced into the thorax. The thoracic wall was stitched. The ventilator was blocked to full fill the lung. The chest muscle and skin was stitched after the air in the thoracic cavity was gently squeezed out. The ventilators were removed from the rats until constant spontaneous respiration resumed.
  • The cardiac function of the rats was then examined by echocardiography (Philips Sonos 7500 S4 probe) on the 14th day after ligation. The rats with ejection fraction (hereinafter "EF") values from 30 to 50 percent were separated and grouped (15 rats per group).
  • B. Treating the ligated rats with neuregulin.
  • The rats were weighed on the 15th day after left ventricle coronary ligation to determine the amount of NRG needed. Rats in the vehicle group received 0.4ml/100g (volume/body weight) of vehicle by IV injection. The vehicle was injected once a day for five days, stopped for two days, and then injected for another five days.
  • The IM and IV groups of rats received IM and IV injection of NRG, respectively (the amount of NRG was 149.3U/kg (protein/body weight), the volume was 0.4ml/100g). The NRG was injected once a day for five days, stopped for two days, and then injected for another five days.
  • As discussed further below, the IVGTT group had osmotic pumps (ALZET osmotic pump 2ML1) implanted on the fifth day after grouping. Each pump contained 2 ml of NRG solution, which contained 933.1U of NRG (as a rat now weighed about 250g) and the infusion speed was about 18.7U/kg/h. Thus, the maximum drug concentration compared to about 2.67U/kg by IV injection.
  • After 7 days, cardiac function of all rats was checked again by echocardiography (Philips Sonos 7500 S4 probe). The next day, hemodynamic parameter check and anatomy check were also undertaken to further confirm the cardiac function of the rats.
  • B.1. Transplantation of osmotic pump into rats (all steps must be sterile)
  • 1ml of sterile water and 1ml of sterile 0.9% saline was injected into a vial of NRG (993.1U, 62.5µg) in the hood successively. The NRG solution was drawn into a sterile syringe. A blunt-tipped needle was exchanged for the syringe and the bubble in the syringe was removed. The pump was held upright and the needle was inserted through the small opening at the top of the upright pump until it could go no further. The plunger was pushed slowly to add NRG solution into the pump until the solution began to overflow the pump. The needle was removed and the pump was wiped clean. The transparent cap of the flow moderator was taken off to expose a short stainless steel tube. The steel tube was then inserted into one end of a 5cm PE60 tube. The syringe needle was inserted into another end of the PE60 tube. The plunger of syringe was pushed to add NRG solution to the flow moderator until it was full. The long tube of the flow moderator was then inserted into the pump until its white flange attached to the pump. The needle was drawn out of the flow moderator before soaking the pump in sterile 0.9% saline at 37°C overnight.
  • The rats were anesthetized by Ketamine (as described above). The area between neck and shoulder of the rats was depilated and sanitized. The body was covered with a piece of sterile wet cloth. An incision was then carefully made in the skin between the scapulae to locate and separate the external jugular vein. The distal end of the vein from the heart was ligated. A small hole was made by eye scissors on the wall of the external jugular vein and enlarged by microforceps. The PE60 tube connected to the osmotic pump was inserted 2cm into the vein through the hole. The proximal end of the vein from the heart was then bound with PE60 tube to fix the tube. The distal end of the vein surrounding the PE60 tube was tied tight to further fix the tube. Using a hemostat, a tunnel was formed by blunt separation of the skin from the incision to scapula. A pocket was finally made on the back of the rat in the midscapular region by spreading the skin further. The pump was slid through the tunnel into the pocket with the flow moderator pointing away from the incision. The skin incision was then closed with a suture. The rats were put back into the animal room after revival and were fed as usual.
  • C. Experimental Results
  • The function of MI heart following NRG infusion by IVGTT and IV is shown in Table 1 below. In Table 1 "IVS", "LVEDD" "PW", "LVESD", "EF", "FS" and "CC" stand for interventricular septum, left ventricle end diastolic dimension, posterior wall thickness, left ventricle end systolic dimension, ejection fraction, fractional shortening and cardiac cycle, respectively. Here EF and FS reflect the contractility of heart, especially for left ventricle. EF = end diastolic volume - end systolic volume / end diastolic volume
    Figure imgb0001
    FS = end diastolic dimension - end systolic dimension / end diastolic dimension
    Figure imgb0002
  • In Table 1, P <0.01 for LVEDD, LVESD, EF and FS in NGTT or IV group compared with their counterparts in the vehicle group, indicating highly significant difference. Table 1, cardiac function of MI rats after NRG infusion by IVGTT and IV
    IVS LVEDD PW LVESD EF FS CC
    cm cm cm cm % % ms
    Vehicle 0.168±0.005 0.952±0.082 0.173±0.009 0.819±0.107 34.3±5.0 14.5±2.4 162.5±23.1
    IVGTT 0.169±0.007 0.857±0.093 0.190±0.013 0.644±0.061 54.6±5.4 25.2±3.0 173.1±22.5
    IV 0.177±0.027 0.912±0.081 0.189±0.013 0.759±0.099 40.5±8.9 17.5±4.6 164.5±18.2
  • NRG infused by osmotic pump dramatically increased the cardiac function of MI rats compared to the IV group. Particularly, the EF value - a measurement of the heart's blood pumping efficiency that can be used to estimate the function of the left ventricle - in the IVGTT group was 59.18% higher than that of the vehicle group, and 34.81% higher than the IV group. Additionally, the FS value - also a way of measuring left ventricle performance - of the IVGTT group was 73.79% higher than that of the vehicle group, and 44.0% higher than the IV group. These results show that extended release of NRG is more effective than conventional IV injection for improving cardiac function.
  • Surprisingly, NRG infused by osmotic pump not only greatly increased the cardiac function of MI rats compare with the IV group, but also reduced the interior diameter of the left ventricle. Specifically, the average Left Ventricle End Diastolic Dimension (hereinafter referred to as "LVEDD") of the IVGTT group was 9.98% smaller than that of the vehicle group, and 6.03% smaller than the IV group. Additionally, the Left Ventricle End Systolic Dimension (hereinafter referred to as "LVESD") of the IVGTT group was 21.37% smaller than that of the vehicle group, and 15.15% smaller than the IV group. These results show that administering NRG constantly can reduce left ventricular volume and mass, thereby improving left ventricular health and performance.
  • EXAMPLE 3 Heart function of myocardial infarcted rats after neuregulin was constantly intravenously infused by syringe pump (Zhejiang University Medical Instrument Co. LTD. WZS 50-F2)
  • In this example, syringe pump is used for extended release of neuregulin in human patients. Syringe pump can pump the solution continuously at certain speed into the bloodstream through a needle injected into the vein in rat tail. For syringe pump, it's easy to control the infusion time and speed. Neuregulin was intravenously infused by syringe pump at different speed for different time per day into MI rats to better time period and speed for treatment.
  • Grouped MI rats was treated by intravenous injection of 4ml/kg (volume/body weight) vehicle everyday for 10 days (group A); or intravenous injection of 10µg/kg neuregulin (2.5µg/ml) everyday for 10 days (group B); or intravenous syringe pump infusion of neuregulin (0.625µg/ml) at 1.25µg/kg/h with 4 hours per day for 10 days (group C); or intravenous syringe pump infusion of neuregulin (1.25µg/ml) at 2.5µg/kg/h with 4 hours per day for 10 days (group D); or intravenous syringe pump infusion of neuregulin (0.625µg/ml) at 0.625µg/kg/h with 8 hours per day for 10 days (group E); or intravenous syringe pump infusion of neuregulin (1.25µg/ml) at 1.25µg/kg/h with 8 hours per day for 10 days (group F). Echocardiography was then performed for all groups to examine the function of heart. Table 2, echocardiography data for MI rats after intravenous syringe pump infusion (ISPI) or IV injection of NRG
    IVS LVEDD PW LVESD EF FS HR
    cm cm cm cm % % /min
    A vehicle 0.057±0.003 0.947±0.041 0.142±0.013 0.811±0.047 34.5±3.3 14.4±1.6 418±51
    B IV 0.060±0.005 0.924±0.060 0.164±0.016 0.770±0.057 41.5±2.6 17.8±1.6 332±52
    C ISPI 1.25µg/kg/h 4h/day 0.059±0.005 0.935±0.050 0.156±0.013 0.779±0.067 41.2±5.7 17.7±2.8 395±30
    D ISPI 2.5µg/kg/h 4h/day 0.061±0.004 0.943±0.058 0.160±0.015 0.762±0.055 43.7±5.4 19.0±2.9 391±41
    E ISPI 0.625µg/kg/ h 8h/day 0.062±0.006 0.941±0.061 0.164±0.011 0.742±0.079 47.4±8.6 21.1±4.5 391±48
    F ISPI 1.25µg/kg/h 8h/day 0.061±0.004 0.966±0.038 0.166±0.019 0.766±0.045 47.2±4.2 20.8±2.5 364±33
  • P < 0.01 for LVEDD, LVEDD, EGF and FS in any of ISPI or IV group compared with their counterparts in the vehicle group, indicating highly significant difference. HR means heart rate.
  • As shown in table 2, compared with the vehicle group, neuregulin by IV (B group) enhanced the EF value of MI rats by 20.29%, intravenous syringe pump infusion for 4h/day (C, D group) was just as effective as IV, while neuregulin by intravenous syringe pump infusion for 8h/day (E, F group) enhanced the EF value by around 37.10%. At the same time, compared with the vehicle group, neuregulin by IV injection (B group) enhanced the FS value of MI rats by 23.61%, intravenous syringe pump infusion for 4h/day (C, D group) was as effective as IV, while neuregulin by intravenous syringe pump infusion for 8h/day (E, F group) enhanced the FS value by around 45.49%. Surprisingly, although MI rats in group E received only half amount of neuregulin for group F, the EF or FS value is nearly the same. The results showed that after neuregulin was continuously intravenously infused by syringe pump for 8 or more hours per day it could enhance the cardiac function.
  • EXAMPLE 4 The cardiac function of MI rats after extended hypodermic infusion of NRG by osmotic pump
  • Left ventricle coronary artery ligation and transplantation of osmotic pump into rats was performed in the same way as in example 2, except the amount of NRG injected into the pump was 1791.3U (125 µg), and the pump was embedded without a tube connected to the vein to make NRG infusion hypodermic. The infusion speed is 37.33U/kg/h.
  • IV infusion was started at the same time as extended hypodermic infusion so the IV group was treated with NRG for 7 days. The amount of NRG for the IV group was also changed to 223.95U/kg.
  • The function of MI heart following NRG infusion by extended hypodermic and IV is shown in Table 3. In Table 3, P< 0.01 for LVEDD, LVESD, EF and FS in the IVGTT and the IV group compared with their counterparts in the vehicle group, indicating a highly significant difference. Table 3, cardiac function of MI rats after extended hypodermic (EHI) and IV infusion of NRG
    IVS LVEDD PW LVESD EF FS CC
    cm cm cm cm % % ms
    Vehicle 0.174±0.005 1.02±0.077 0.185±0.012 0.876±0.098 33.9±7.9 14.3±3.8 153±19
    EHI 0.177±0.006 0.908±0.079 0.209±0.023 0.712±0.091 48:4±9.3 21.7±5.1 153±11
    IV 0.171±0.007 1.013±0.111 0.188±0.010 0.874±0.124 33.9±6.8 14.3±3.3 157±15
  • Table 3 shows that extended hypodermic infusion of NRG significantly increased the cardiac function of MI rats compared to the IV and vehicle groups. Compared to vehicle group, extended hypodermic infusion of NRG enhanced the EF value of MI hearts 42.77%, the FS value 51.75%. As discussed above, the EF and FS values are ways of measuring the heart's blood pumping efficiency and can be used to estimate the function of the left ventricle. Thus, these results show that extended release of NRG is much more effective than conventional IV injection for improving cardiac function.
  • Extended hypodermic infusion of NRG also reduced interior diameter of the left ventricle. Specifically, the LVEDD of MI hearts decreased 10.98% and the LVESD decreased 18.72% compare to vehicle group. IV injection of NRG in this experiment did not have an obvious effect on the cardiac function of MI heart compare to vehicle. The results show that extended hypodermic infusion of NRG can reduce left ventricular volume and mass, thereby improving left ventricular health and performance, which suggests that it may also be used as a treatment for heart failure.
  • EXAMPLE 5 Heart function of myocardial infarcted rats after neuregulin was constantly hypodermically infused by syringe_pump
  • Neuregulin was further infused by syringe pump at different speed for different time per day into MI rats.
  • Grouped MI rats was treated by intravenous injection of 4ml/kg (volume/body weight) vehicle everyday for 10 days (group A); or intravenous injection of 10µg/kg neuregulin (2.5µg/ml) everyday for 10 days (group B); or hypodermic injection (HI) of 10µg/kg neuregulin (2.5µg/ml) everyday for 10 days (group C); hypodermic syringe pump infusion of neuregulin (1.25µg/ml) at 2.5µg/kg/h with 4 hours per day for 10 days (group D); or hypodermic syringe pump infusion of neuregulin (1.11 µg/ml) at 1.67µg/kg/h with 6 hours per day for 10 days (group E); or hypodermic syringe pump infusion of neuregulin (1.25µg/ml) at 1.25µg/kg/h with 8 hours per day for 10 days (group F). Echocardiography was then performed for all groups to examine the function of heart. Table 4, echocardiography data for MI rats after hypodermic syringe pump infusion (HSPI) or IV injection of NRG
    IVS LVEDD PW LVESD EF FS HR
    cm cm cm cm % % /min
    A vehicle 0.060±0.007 0.906±0.107 0.151±0.027 0.757±0.130 39.3±10.8 16.9±6.1 388±33
    B IV 0.063±0.004 0.812±0.045 0.159±0.010 0.726±0.047 43.4±2.8 18.8±1.4 383±33
    C HI 0.063±0.003 0.909±0.054 0.163±0.011 0.744+0.048 42.1±3.7 18.1±1.9 390±40
    D HSPI 2.5µg/kg/h 4h/day 0.065±0.007 0.933±0.055 0.160±0.016 0.754±0.069 44.2±6.5 19.3±3.4 385±32
    E HSPI 1.67µg/kg/h 6h/day 0.067±0.003 0.880±0.073 0.168±0.019 0.693±0.076 48.3±6.0 21.4±3.5 404±38
    F HSPI 1.25µg/kg/h 8h/day 0.066±0.005 0.899±0.056 0.168±0.014 0.709±0.098 472±11.8 21.3±8.2 377±44
  • P<0.01 for LVEDD, LVEDD, EF and FS in any of HSPI, HI or IV group compared with their counterparts in the vehicle group, indicating highly significant difference. HR means heart rate.
  • As shown in table 4, compared with the vehicle group, neuregulin by IV (B group) enhanced the EF value of MI rats by 10.43%, neuregulin by hypodermic injection (C group) enhanced the EF value of MI rats by 7.12%, while neuregulin by hypodermic syringe pump infusion for 4h/day (D group) enhanced the EF value by 12.47%, neuregulin by hypodermic syringe pump infusion for 6h/day (E group) made the EF value jump to 22.90%, neuregulin by hypodermic syringe pump infusion for 8h/day (E group) also raised the EF value by 20.10%. At the same time, compared with the vehicle group, neuregulin by IV (B group) enhanced the FS value of MI rats by 11.24%, neuregulin by hypodermic injection (C group) enhanced the FS value of MI rats by 7.10%, while neuregulin by hypodermic syringe pump infusion for 4h/day (D group) enhanced the FS value by 14.20%, neuregulin by hypodermic syringe pump infusion for 6h/day (E group) made the FS value jump to 26.63%, neuregulin by hypodermic syringe pump infusion for 8h/day (E group) also raised the FS value by 26.04%. The results showed that after neuregulin was continuously hypodermically infused by syringe pump for 6 or more hours per day could it increased the cardiac function dramatically.
  • EXAMPLE 6 PEG coupling of NRG and activity of PEG coupled NRG A, PEG coupling and isolation of PEG coupled NRG
  • PEG (mPEG-SPA-5000, NEKTAR) was added into 10ml 20mM PBS (pH 8.0) containing 1 mg/ml NRG (PEG:NRG=1:1, molar ratio) and mixed quickly, and the mixture was gently stirred at room temperature for 30min, then certain amount of glacial acetic acid was added to stop coupling reaction. The mixture was then loaded onto a gel filtration column (S100, Pharmacia) to separate the components. Each peak fraction was collected and its sample was prepared for SDS-PAGE. After electrophoresis, the gel was stained by BaI2 and Coomassie brilliant blue sequentially to detect PEG and NRG separately.
  • As shown in figure 2 for BaI2 stained gel, the mixture contains PEG monomer, NRG-monoPEG, NRG-diPEG and NRG-polyPEG. After the mixture was loaded onto a S100 gel filtration column, the components were well separated into NRG-polyPEG and NRG-diPEG (peak1), NRG-monoPEG and PEG (peak2).
  • Coomassie stained gel in figure 3 further confirmed that peak1 and peak2 contain NRG which was coupled to PEG, while peak3 contains only NRG.
  • B, measuring activity of PEG coupled NRG
  • MCF-7 cells was harvested, counted, pelleted and resuspended into DMEM (with 10% serum and 9µg/ml insulin) at 5×104 cells/ml. 100µl cell suspension was added to each well of 96 well plate and the plate was incubated at 37°C overnight. The cells were then washed 3 times with PBS and grew in serum free DMEM for another 24 hours.
  • ErbB2 antibody H4 (Zensun, anti-ErbB2 monoclonal antibody) was diluted to 6µg/ml by coating buffer (50mM Na2CO3-NaHCO3, pH9.6), and added to 96 well plate 50µl/well. The plate was left at 4°C overnight to coat with antibody.
  • DMEM was sucked away from the starved MCF-7 cells, and 100µl serial dilutions of NRG, NRG-monoPEG or NRG-diPEG in DMEM was added to each well separately. DMEM was added to two wells as blank. The plate was incubated at 37°C for 20min. The cells were washed once with PBS before adding 100µl/well lysis buffer (50mM Hepes, pH 8.0,150mM NaCl, 2mM sodium orthovanadate, 0.01% thimerosal, 1% Triton X-100 and one protease inhibitor cocktail tablet per 25ml solution) and lysing at 4°C for 30min. The plate was then shaken gently to completely lyse and remove cells from the plate and centrifugated at 15000rpm for 15min.
  • The plate with coating antibody was washed five times with washing buffer (10mM PBS, pH7.4, 0.05% Tween 20) before adding 200µl well of 5% nonfat milk in washing buffer. The plate was incubated at 37°C for 2 hours before washed again 3 times with washing buffer.
  • A 90µl solution of lysed cells was drawn from each well in culture plate and transferred to corresponding well in coated plate. Following incubation at 37°C for 1 hour, the coated plate with cell lysis was washed again 5 times with washing buffer and treated with 100µl suitable concentration of horseradish peroxidase (HRP) conjugated anti-phosphotyrosine monoclonal antibody (Santa Cruz Biotechnology) at 37°C for 1 hour. After the plate was washed again 5 times with washing buffer, 100µl freshly prepared HRP substrate solution [50mM citric acid, 100mM Na2PO4, pH 5.0, 0.2mg/ml 3,3',5,5'-tetramethylbenzidine (TMB), 0.003% H2O2] was added to each well before the plate was incubated at 37°C for 10min. Finally 50µl of 2N H2SO4: was added to each well to destroy HRP activity. The OD value at 450nm for each well was read on a microplate reader (BIO-RAD Model 550), and EC50 was the concentration of NRG which achieved 50% of maximum OD value. The lower the EC50, the higher the activity.
  • The EC50 of NRG, NRG-monoPEG and NRG-diPEG was shown in Table 5. Table 5, EC50 of NRG, NRG-monoPEG and NRG-diPEG
    sample EC50 (µg/ml)
    NRG 0.070
    NRG-monoPEG 0.070
    NRG-diPEG 0.098
  • From table 5, we can see clearly that EC50 of NFG-monoPEG is the same as that of NRG, while EC50 of NRG-diPEG is 40% higher. This means that NRG-monoPEG has the same activity as NRG in vitro, but the activity of NRG-diPEG is 40% lower.
  • EXAMPLE 7 Extended release of neuregulin reduces side effects of neuregulin administration
  • This examples shows that compared with long time or high dose administration, extended release of neuregulin can reduce side effects, such as gastrointestinal disorder or pericardial effusion, associated with neuregulin administration.
  • NRG-1β was administered intravenously by syringe pump to two groups of monkeys, each consisting of twenty four healthy rhesus monkeys (twelve male and twelve female, weighing about 5-7kg). Group I was infused with NRG-1β for twelve hours a day for fourteen days, at the speed of 1 ug/kg/hr. No Side effect was observed in this group. Group II was infused for twenty four hours a day for fourteen days, at the speed of 1ug/kg/hr. In Group II, about 3-5 ml pericardial effusion in the heart of monkeys was observed.
  • Two groups of healthy individuals were administered the same amount of NRG-1β per day for 10 days. Eight individuals in Group I, were infused with NRG-1β for four hours each day for ten days at speed of 0.3 µg/kg/hr. In this group, each individual on average experienced gastrointestinal disorder about two times during the ten-day period. Six individuals were infused with NRG-1β for two hours each day for ten days at speed of 0.6 µg/kg/hr. In Group II, each individual on average experienced gastrointestinal disorder about five times during the ten-day period.
  • These results show that extended release of neuregulin can reduce adverse side effects associated with long time or high dose neuregulin administration. These results suggest that intravenously or hypodermically infusion for hsoert time or lower doage per day could reduce the side effects of 24-hour neuregulin infusion.
  • EXAMPLE 8 Gene expression by extended released NRG in the left ventricle of myocardial infarcted rat
  • In this example, myocardial infarcted rats were infused with NRG-1β and gene expression pattern in the left ventricle of these rats was analyzed by microarray. Compare with myocardial infarcted rats infused with vehicle, rats infused with NRG have different gene expression pattern. After extended release of NRG, thymosin beta like protein mRNA level increased 3.10 times; defensin beta.1 mRNA level increased 2.87 times; growth associated protein mRNA level increased 2.16 times; mRNA level of thymosin beta 4, Laminin gamma 1, myocardin, PI3K gamma regulatory subunit almost all doubled, while mRNA level of Elastin and PI3K gamma was nearly the same as before. It shows that neuregulin changes the expression level of various proteins in heart.
  • SEQUENCE LISTING
    • <110> Zensun (Shanghai) Science and Technology Inc. Zhou, Mingdong
    • <120> EXTENDED RELEASE OF NEUREGULIN FOR IMPROVED CARDIAC FUNCTION
    • <130> 11748-009-228
    • <140> To be Assigned
      <141> 2006-12-29
    • <150> US 60/755,124
      <151> 2005-12-30
    • <150> US 60/758,626
      <151> 2006-01-13
    • <160> 2
    • <170> FastSEQ for Windows Version 4.0
    • <210> 1
      <211> 61
      <212> PRT
      <213> Homo sapiens
    • <400> 1
      Figure imgb0003
    • <210> 2
      <211> 183
      <212> DNA
      <213> Homo sapiens
    • <400> 2
      Figure imgb0004

Claims (6)

  1. Neuregulin 1 or a functional fragment thereof comprising an epidermal growth factor-like domain of Neuregulin 1 for use in a method of preventing, treating or delaying heart failure in a mammal, wherein the method comprises releasing Neuregulin 1 to the mammal by extended release means over a period of between 4 hours and 12 hours per day, wherein the extended release means comprise an osmotic pump or syringe pump.
  2. The Neuregulin 1 or fragment thereof for use of claim 1, wherein said Neuregulin 1 is released at a dose of about 1 U/kg to 5000 U/kg of body weight, or 10 U/kg to 1000 U/kg of body weight of the mammal.
  3. The Neuregulin 1 or fragment thereof for use of claim 1, wherein said Neuregulin 1 is released at a dose of about 1 U/day to 10,000 U/day.
  4. The Neuregulin 1 or fragment thereof for use of claim 1, wherein said Neuregulin 1 is released at a dose of about 0,001 mg/day to 1000 mg/day.
  5. The Neuregulin 1 or fragment thereof for use of any one of claims 1-4, wherein said Neuregulin 1 fragment comprises the amino acid sequence of SEQ ID NO:1.
  6. The Neuregulin 1 or fragment thereof for use of any one of claims 1-5, wherein the mammal is a human.
EP06840724.6A 2005-12-30 2006-12-29 Extended release of neuregulin for improved cardiac function Active EP1981525B1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
EP15151306.6A EP2918283B1 (en) 2005-12-30 2006-12-29 Extended release of neuregulin for improved cardiac function
EP18153913.1A EP3363455A1 (en) 2005-12-30 2006-12-29 Extended release of neuregulin for improved cardiac function

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US75512405P 2005-12-30 2005-12-30
US75862606P 2006-01-13 2006-01-13
PCT/CN2006/003694 WO2007076701A1 (en) 2005-12-30 2006-12-29 Extended release of neuregulin for improved cardiac function

Related Child Applications (2)

Application Number Title Priority Date Filing Date
EP15151306.6A Division EP2918283B1 (en) 2005-12-30 2006-12-29 Extended release of neuregulin for improved cardiac function
EP18153913.1A Division EP3363455A1 (en) 2005-12-30 2006-12-29 Extended release of neuregulin for improved cardiac function

Publications (3)

Publication Number Publication Date
EP1981525A1 EP1981525A1 (en) 2008-10-22
EP1981525A4 EP1981525A4 (en) 2009-11-11
EP1981525B1 true EP1981525B1 (en) 2015-01-21

Family

ID=38227911

Family Applications (3)

Application Number Title Priority Date Filing Date
EP06840724.6A Active EP1981525B1 (en) 2005-12-30 2006-12-29 Extended release of neuregulin for improved cardiac function
EP15151306.6A Active EP2918283B1 (en) 2005-12-30 2006-12-29 Extended release of neuregulin for improved cardiac function
EP18153913.1A Withdrawn EP3363455A1 (en) 2005-12-30 2006-12-29 Extended release of neuregulin for improved cardiac function

Family Applications After (2)

Application Number Title Priority Date Filing Date
EP15151306.6A Active EP2918283B1 (en) 2005-12-30 2006-12-29 Extended release of neuregulin for improved cardiac function
EP18153913.1A Withdrawn EP3363455A1 (en) 2005-12-30 2006-12-29 Extended release of neuregulin for improved cardiac function

Country Status (9)

Country Link
US (3) US20070190127A1 (en)
EP (3) EP1981525B1 (en)
JP (3) JP5738516B2 (en)
AU (1) AU2006332340B2 (en)
BR (1) BRPI0620819A2 (en)
CA (2) CA2634974A1 (en)
ES (2) ES2664086T3 (en)
RU (1) RU2457854C2 (en)
WO (1) WO2007076701A1 (en)

Families Citing this family (28)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AUPP785098A0 (en) * 1998-12-21 1999-01-21 Victor Chang Cardiac Research Institute, The Treatment of heart disease
AU2002304965A1 (en) * 2002-05-24 2003-12-12 Zensun (Shanghai) Sci-Tech.Ltd Neuregulin based methods and compositions for treating viral myocarditis and dilated cardiomyopathy
US9034316B2 (en) * 2006-10-24 2015-05-19 Amorcyte, Llc Infarct area perfusion-improving compositions and methods of vascular injury repair
US8637005B2 (en) 2005-11-07 2014-01-28 Amorcyte, Inc. Compositions and methods of vascular injury repair
US20110076255A1 (en) 2005-11-07 2011-03-31 Pecora Andrew L Compositions and methods for treating progressive myocardial injury due to a vascular insufficiency
EP1981525B1 (en) 2005-12-30 2015-01-21 Zensun (Shanghai) Science and Technology Limited Extended release of neuregulin for improved cardiac function
US20090156488A1 (en) * 2007-09-12 2009-06-18 Zensun (Shanghai) Science & Technology Limited Use of neuregulin for organ preservation
US20100256066A1 (en) * 2007-11-16 2010-10-07 Proteosys Ag Active soluble post-translationally modified neuregulin isoforms
EP2320933B1 (en) 2008-07-17 2017-12-27 Acorda Therapeutics, Inc. Therapeutic dosing of a neuregulin or a subsequence thereof for treatment or prophylaxis of heart failure
CN102232084B (en) 2008-11-28 2014-05-28 上海泽生科技开发有限公司 Neuregulin peptides and their use
CN102231987A (en) * 2008-11-28 2011-11-02 上海泽生科技开发有限公司 Neuregulin and cardiac stem cells
ES2748886T3 (en) 2009-06-09 2020-03-18 Zensun Shanghai Science & Tech Co Ltd Neuregulin-based methods for the treatment of heart failure
WO2010142141A1 (en) * 2009-06-09 2010-12-16 Zensun (Shanghai) Science & Technology Limited Neuregulin based methods for treating heart failure
EP2493495A4 (en) * 2009-07-10 2013-08-21 Univ Northwestern Cardioprotective role of hepatic cells and hepatocyte secretory factors in myocardial ischemia
WO2011011388A2 (en) * 2009-07-22 2011-01-27 Children's Medical Center Corporation Neuregulin induced regeneraton of heart muscle muscle
JP6096262B2 (en) 2009-08-25 2017-03-15 ゼンサン (シャンハイ) サイエンス アンド テクノロジー,シーオー.,エルティーディー. Treatment of heart failure based on neuregulin
EP2491114B8 (en) * 2009-10-23 2017-01-25 Amorcyte, Inc. Compositions and uses for treating progressive myocardial injury due to a vascular insufficiency
AR079814A1 (en) 2009-12-31 2012-02-22 Otsuka Pharma Co Ltd HETEROCICLICAL COMPOUNDS, PHARMACEUTICAL COMPOSITIONS THAT CONTAIN THEM AND THEIR USES
CN102139095A (en) 2010-01-29 2011-08-03 上海泽生科技开发有限公司 Method for applying neuregulin to prevent, treat or delay cardiac ischemia reperfusion injuries, and composition for preventing, treating or delaying cardiac ischemia reperfusion injuries
JP6121658B2 (en) * 2011-06-29 2017-04-26 大塚製薬株式会社 Therapeutic compounds and related methods of use
WO2013053076A1 (en) 2011-10-10 2013-04-18 Zensun (Shanghai)Science & Technology Limited Compositions and methods for treating heart failure
US9533010B2 (en) 2011-10-31 2017-01-03 Amorcyte, Llc Compositions and methods for treating progressive myocardial injury due to a vascular insufficiency
CN104884077B (en) * 2012-10-08 2019-02-12 上海泽生科技开发股份有限公司 The component and method for treating diabetic's heart failure
BR112015029293A2 (en) * 2013-05-22 2018-04-24 Zensun Shanghai Science & Tech Ltd method and kit for preventing, treating or delaying a cardiovascular disease or disorder in a mammal
CN110946993A (en) * 2014-01-03 2020-04-03 上海泽生科技开发股份有限公司 Formula of neuregulin preparation
CN105497876B (en) 2014-09-24 2021-01-15 上海泽生科技开发股份有限公司 Methods and compositions for the prevention, treatment or delay of cardiac ventricular arrhythmias with neuregulin
CN105561298A (en) 2014-10-17 2016-05-11 上海泽生科技开发有限公司 Method for preventing, treating or delaying ejection fraction reserved cardiac failure by means of neuregulin and composition
AR121035A1 (en) 2019-04-01 2022-04-13 Lilly Co Eli NEUREGULIN-4 COMPOUNDS AND METHODS OF USE

Family Cites Families (153)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3536809A (en) 1969-02-17 1970-10-27 Alza Corp Medication method
US3598123A (en) 1969-04-01 1971-08-10 Alza Corp Bandage for administering drugs
US3773919A (en) 1969-10-23 1973-11-20 Du Pont Polylactide-drug mixtures
US3845770A (en) 1972-06-05 1974-11-05 Alza Corp Osmatic dispensing device for releasing beneficial agent
US3916899A (en) 1973-04-25 1975-11-04 Alza Corp Osmotic dispensing device with maximum and minimum sizes for the passageway
US4179337A (en) 1973-07-20 1979-12-18 Davis Frank F Non-immunogenic polypeptides
GB1478759A (en) * 1974-11-18 1977-07-06 Alza Corp Process for forming outlet passageways in pills using a laser
US4002531A (en) 1976-01-22 1977-01-11 Pierce Chemical Company Modifying enzymes with polyethylene glycol and product produced thereby
US4008719A (en) 1976-02-02 1977-02-22 Alza Corporation Osmotic system having laminar arrangement for programming delivery of active agent
US4263428A (en) 1978-03-24 1981-04-21 The Regents Of The University Of California Bis-anthracycline nucleic acid function inhibitors and improved method for administering the same
US4200098A (en) 1978-10-23 1980-04-29 Alza Corporation Osmotic system with distribution zone for dispensing beneficial agent
JPS6023084B2 (en) 1979-07-11 1985-06-05 味の素株式会社 blood substitute
ATE12348T1 (en) 1980-11-10 1985-04-15 Gersonde Klaus Prof Dr PROCESS FOR THE PRODUCTION OF LIPID VESICLES BY ULTRASONIC TREATMENT, APPLICATION OF THE PROCESS AND DEVICE FOR CARRYING OUT THE PROCESS.
IE52535B1 (en) 1981-02-16 1987-12-09 Ici Plc Continuous release pharmaceutical compositions
US4485045A (en) 1981-07-06 1984-11-27 Research Corporation Synthetic phosphatidyl cholines useful in forming liposomes
US4640835A (en) 1981-10-30 1987-02-03 Nippon Chemiphar Company, Ltd. Plasminogen activator derivatives
JPS58118008A (en) 1982-01-06 1983-07-13 Nec Corp Data processor
EP0088046B1 (en) 1982-02-17 1987-12-09 Ciba-Geigy Ag Lipids in the aqueous phase
DE3218121A1 (en) 1982-05-14 1983-11-17 Leskovar, Peter, Dr.-Ing., 8000 München Pharmaceutical compositions for tumour treatment
EP0102324A3 (en) 1982-07-29 1984-11-07 Ciba-Geigy Ag Lipids and surfactants in an aqueous medium
US4542025A (en) 1982-07-29 1985-09-17 The Stolle Research And Development Corporation Injectable, long-acting microparticle formulation for the delivery of anti-inflammatory agents
US4544545A (en) 1983-06-20 1985-10-01 Trustees University Of Massachusetts Liposomes containing modified cholesterol for organ targeting
JPS607934A (en) 1983-06-29 1985-01-16 Dai Ichi Seiyaku Co Ltd Preparation of liposome
HUT35524A (en) 1983-08-02 1985-07-29 Hoechst Ag Process for preparing pharmaceutical compositions containing regulatory /regulative/ peptides providing for the retarded release of the active substance
DE3486459D1 (en) 1983-09-26 1997-12-11 Udo Dr Med Ehrenfeld Means and product for the diagnosis and therapy of tumors and for the treatment of weaknesses in cellular and humoral immune defense
DE3474511D1 (en) 1983-11-01 1988-11-17 Terumo Corp Pharmaceutical composition containing urokinase
FR2621486B1 (en) 1987-10-07 1990-01-26 Synthelabo USE OF INDOLE PYRROLO (3,2,1-HI) DERIVATIVES FOR OBTAINING A MEDICAMENT FOR THE TREATMENT OF DIABETES
US4496689A (en) 1983-12-27 1985-01-29 Miles Laboratories, Inc. Covalently attached complex of alpha-1-proteinase inhibitor with a water soluble polymer
US5231112A (en) 1984-04-12 1993-07-27 The Liposome Company, Inc. Compositions containing tris salt of cholesterol hemisuccinate and antifungal
US5916588A (en) 1984-04-12 1999-06-29 The Liposome Company, Inc. Peptide-containing liposomes, immunogenic liposomes and methods of preparation and use
ES8702440A1 (en) 1984-10-04 1986-12-16 Monsanto Co Prolonged release of biologically active somatotropins.
IE58110B1 (en) 1984-10-30 1993-07-14 Elan Corp Plc Controlled release powder and process for its preparation
DE3675588D1 (en) 1985-06-19 1990-12-20 Ajinomoto Kk HAEMOGLOBIN TIED TO A POLY (ALKENYLENE OXIDE).
US5059421A (en) 1985-07-26 1991-10-22 The Liposome Company, Inc. Preparation of targeted liposome systems of a defined size distribution
MX9203291A (en) 1985-06-26 1992-08-01 Liposome Co Inc LIPOSOMAS COUPLING METHOD.
US5171578A (en) 1985-06-26 1992-12-15 The Liposome Company, Inc. Composition for targeting, storing and loading of liposomes
US4885172A (en) 1985-06-26 1989-12-05 The Liposome Company, Inc. Composition for targeting, storing and loading of liposomes
US4684479A (en) 1985-08-14 1987-08-04 Arrigo Joseph S D Surfactant mixtures, stable gas-in-liquid emulsions, and methods for the production of such emulsions from said mixtures
US6759057B1 (en) 1986-06-12 2004-07-06 The Liposome Company, Inc. Methods and compositions using liposome-encapsulated non-steroidal anti-inflammatory drugs
US4791192A (en) 1986-06-26 1988-12-13 Takeda Chemical Industries, Ltd. Chemically modified protein with polyethyleneglycol
IE60901B1 (en) 1986-08-21 1994-08-24 Vestar Inc Improved treatment of systemic fungal infections with phospholipid particles encapsulating polyene antifungal antibiotics
US5811128A (en) 1986-10-24 1998-09-22 Southern Research Institute Method for oral or rectal delivery of microencapsulated vaccines and compositions therefor
US6406713B1 (en) 1987-03-05 2002-06-18 The Liposome Company, Inc. Methods of preparing low-toxicity drug-lipid complexes
US4897268A (en) 1987-08-03 1990-01-30 Southern Research Institute Drug delivery system and method of making the same
WO1989001489A1 (en) 1987-08-10 1989-02-23 Commonwealth Scientific And Industrial Research Or Control of angiogenesis and compositions and methods therefor
AU598958B2 (en) 1987-11-12 1990-07-05 Vestar, Inc. Improved amphotericin b liposome preparation
US4904584A (en) 1987-12-23 1990-02-27 Genetics Institute, Inc. Site-specific homogeneous modification of polypeptides
US5073543A (en) 1988-07-21 1991-12-17 G. D. Searle & Co. Controlled release formulations of trophic factors in ganglioside-lipsome vehicle
JPH02226533A (en) 1989-02-27 1990-09-10 Mitsubishi Electric Corp Information recording medium
IT1229203B (en) * 1989-03-22 1991-07-25 Bioresearch Spa USE OF 5 METHYLTHETRAHYDROPHOLIC ACID, 5 FORMYLTHETRAHYDROPHOLIC ACID AND THEIR PHARMACEUTICALLY ACCEPTABLE SALTS FOR THE PREPARATION OF PHARMACEUTICAL COMPOSITIONS IN THE FORM OF CONTROLLED RELEASE ACTIVE IN THE THERAPY OF MENTAL AND ORGANIC DISORDERS.
EP0471036B2 (en) * 1989-05-04 2004-06-23 Southern Research Institute Encapsulation process
PH30995A (en) * 1989-07-07 1997-12-23 Novartis Inc Sustained release formulations of water soluble peptides.
US5120548A (en) 1989-11-07 1992-06-09 Merck & Co., Inc. Swelling modulated polymeric drug delivery device
US5733566A (en) * 1990-05-15 1998-03-31 Alkermes Controlled Therapeutics Inc. Ii Controlled release of antiparasitic agents in animals
US6517859B1 (en) 1990-05-16 2003-02-11 Southern Research Institute Microcapsules for administration of neuroactive agents
US5215680A (en) 1990-07-10 1993-06-01 Cavitation-Control Technology, Inc. Method for the production of medical-grade lipid-coated microbubbles, paramagnetic labeling of such microbubbles and therapeutic uses of microbubbles
GB9107566D0 (en) 1991-04-10 1991-05-29 Ludwig Inst Cancer Res Glial mitogenic factors,their preparation and use
US5716930A (en) 1991-04-10 1998-02-10 Ludwig Institute For Cancer Research Glial growth factors
US7115554B1 (en) 1993-05-06 2006-10-03 Acorda Therapeutics, Inc. Methods of increasing myotube formation or survival or muscle cell mitogenesis differentiation or survival using neuregulin GGF III
US5530109A (en) 1991-04-10 1996-06-25 Ludwig Institute For Cancer Research DNA encoding glial mitogenic factors
IL101943A0 (en) * 1991-05-24 1992-12-30 Genentech Inc Structure,production and use of heregulin
US5834229A (en) * 1991-05-24 1998-11-10 Genentech, Inc. Nucleic acids vectors and host cells encoding and expressing heregulin 2-α
US5367060A (en) 1991-05-24 1994-11-22 Genentech, Inc. Structure, production and use of heregulin
US5580578A (en) 1992-01-27 1996-12-03 Euro-Celtique, S.A. Controlled release formulations coated with aqueous dispersions of acrylic polymers
US6087323A (en) 1992-04-03 2000-07-11 Cambridge Neuroscience, Inc. Use of neuregulins as modulators of cellular communication
US7037888B1 (en) * 1992-04-03 2006-05-02 Acorda Therapeutics, Inc. Methods for treating muscle diseases and disorders
US5573776A (en) 1992-12-02 1996-11-12 Alza Corporation Oral osmotic device with hydrogel driving member
TW333456B (en) 1992-12-07 1998-06-11 Takeda Pharm Ind Co Ltd A pharmaceutical composition of sustained-release preparation the invention relates to a pharmaceutical composition of sustained-release preparation which comprises a physiologically active peptide.
US5591767A (en) 1993-01-25 1997-01-07 Pharmetrix Corporation Liquid reservoir transdermal patch for the administration of ketorolac
US6090925A (en) 1993-03-09 2000-07-18 Epic Therapeutics, Inc. Macromolecular microparticles and methods of production and use
US5554730A (en) * 1993-03-09 1996-09-10 Middlesex Sciences, Inc. Method and kit for making a polysaccharide-protein conjugate
US5981719A (en) 1993-03-09 1999-11-09 Epic Therapeutics, Inc. Macromolecular microparticles and methods of production and use
ATE258685T1 (en) 1993-03-09 2004-02-15 Baxter Int MACROMOLECULAR MICROPARTICLES AND METHOD FOR THE PRODUCTION THEREOF
US6087324A (en) 1993-06-24 2000-07-11 Takeda Chemical Industries, Ltd. Sustained-release preparation
CA2126619A1 (en) 1993-06-24 1994-12-25 Yasutaka Igari Sustained-release preparation of anti-endothelin substance
US5446090A (en) * 1993-11-12 1995-08-29 Shearwater Polymers, Inc. Isolatable, water soluble, and hydrolytically stable active sulfones of poly(ethylene glycol) and related polymers for modification of surfaces and molecules
IT1270594B (en) 1994-07-07 1997-05-07 Recordati Chem Pharm CONTROLLED RELEASE PHARMACEUTICAL COMPOSITION OF LIQUID SUSPENSION MOGUISTEIN
US5824784A (en) 1994-10-12 1998-10-20 Amgen Inc. N-terminally chemically modified protein compositions and methods
US5770567A (en) * 1994-11-14 1998-06-23 Genentech, Inc. Sensory and motor neuron derived factor (SMDF)
US5932462A (en) * 1995-01-10 1999-08-03 Shearwater Polymers, Inc. Multiarmed, monofunctional, polymer for coupling to molecules and surfaces
JP3342873B2 (en) * 1995-03-10 2002-11-11 ジェネンテク・インコーポレイテッド Receptor activation by gas6
US6750196B1 (en) * 1995-03-27 2004-06-15 Acorda Therapeutics Methods of treating disorders of the eye
US6033660A (en) * 1995-05-10 2000-03-07 Genentech, Inc. Method of treating a nervous system injury with cultured schwann cells
US5721139A (en) * 1995-05-10 1998-02-24 Genentech, Inc. Isolating and culturing schwann cells
US5714385A (en) * 1995-05-10 1998-02-03 Genentech, Inc. Media for culturing schwann cells
ATE268591T1 (en) * 1995-06-27 2004-06-15 Takeda Chemical Industries Ltd METHOD FOR PRODUCING DELAYED RELEASE PREPARATIONS
TW448055B (en) 1995-09-04 2001-08-01 Takeda Chemical Industries Ltd Method of production of sustained-release preparation
US5912326A (en) 1995-09-08 1999-06-15 President And Fellows Of Harvard College Cerebellum-derived growth factors
JP2909418B2 (en) 1995-09-18 1999-06-23 株式会社資生堂 Delayed release microsphere of drug
US5980945A (en) 1996-01-16 1999-11-09 Societe De Conseils De Recherches Et D'applications Scientifique S.A. Sustained release drug formulations
US5968511A (en) 1996-03-27 1999-10-19 Genentech, Inc. ErbB3 antibodies
US6264970B1 (en) 1996-06-26 2001-07-24 Takeda Chemical Industries, Ltd. Sustained-release preparation
ATE290595T1 (en) * 1996-07-12 2005-03-15 Genentech Inc GAMMA-HEREGULIN
ES2208946T3 (en) 1996-08-23 2004-06-16 Sequus Pharmaceuticals, Inc. LIPOSOMES CONTAINING A CISPLATIN COMPOUND.
US6419961B1 (en) * 1996-08-29 2002-07-16 Takeda Chemical Industries, Ltd. Sustained release microcapsules of a bioactive substance and a biodegradable polymer
CA2217134A1 (en) * 1996-10-09 1998-04-09 Sumitomo Pharmaceuticals Co., Ltd. Sustained release formulation
EP0932390A1 (en) 1996-10-11 1999-08-04 Sequus Pharmaceuticals, Inc. Therapeutic liposome composition and method
US6056973A (en) 1996-10-11 2000-05-02 Sequus Pharmaceuticals, Inc. Therapeutic liposome composition and method of preparation
AU725257B2 (en) 1996-10-15 2000-10-12 Transave, Inc. N-acyl phosphatidylethanolamine-mediated liposomal drug delivery
US6361796B1 (en) 1996-10-25 2002-03-26 Shire Laboratories, Inc. Soluble form osmotic dose delivery system
US6110498A (en) 1996-10-25 2000-08-29 Shire Laboratories, Inc. Osmotic drug delivery system
EP0839525B1 (en) 1996-10-31 2004-08-04 Takeda Chemical Industries, Ltd. Sustained-release preparation
US6593290B1 (en) * 1996-11-01 2003-07-15 Genentech, Inc. Treatment of inner ear hair cells
US6156728A (en) 1996-11-01 2000-12-05 Genentech, Inc. Treatment of inner ear hair cells
EP0842657A1 (en) 1996-11-19 1998-05-20 OctoPlus B.V. Microspheres for controlled release and processes to prepare these microspheres
US6395302B1 (en) 1996-11-19 2002-05-28 Octoplus B.V. Method for the preparation of microspheres which contain colloidal systems
EP0946169B1 (en) 1996-12-20 2003-02-26 Takeda Chemical Industries, Ltd. Method of producing a sustained-release preparation
US5891474A (en) 1997-01-29 1999-04-06 Poli Industria Chimica, S.P.A. Time-specific controlled release dosage formulations and method of preparing same
US6136558A (en) 1997-02-10 2000-10-24 Genentech, Inc. Heregulin variants
ATE427353T1 (en) * 1997-02-10 2009-04-15 Genentech Inc HEREGULIN VARIANTS
WO1998055611A1 (en) 1997-06-06 1998-12-10 Regents Of The University Of Michigan Neuregulin response element and uses therefor
US6121415A (en) * 1997-07-09 2000-09-19 Genentech, Inc. ErbB4 receptor-specific neuregolin related ligands and uses therefor
DE19747261A1 (en) * 1997-10-25 1999-04-29 Bayer Ag Single-chamber osmotic pharmaceutical release system
WO1999018976A1 (en) * 1997-10-14 1999-04-22 Cambridge Neuroscience, Inc. Therapeutic methods comprising use of a neuregulin
HUP0100221A3 (en) 1998-01-16 2002-11-28 Takeda Pharmaceutical Sustained release compositions, process for producing the same and utilization thereof
US6613358B2 (en) 1998-03-18 2003-09-02 Theodore W. Randolph Sustained-release composition including amorphous polymer
KR19990085365A (en) * 1998-05-16 1999-12-06 허영섭 Biodegradable polymer microspheres capable of continuously controlled controlled release and preparation method thereof
US6713086B2 (en) 1998-12-18 2004-03-30 Abbott Laboratories Controlled release formulation of divalproex sodium
AUPP785098A0 (en) 1998-12-21 1999-01-21 Victor Chang Cardiac Research Institute, The Treatment of heart disease
US6248363B1 (en) 1999-11-23 2001-06-19 Lipocine, Inc. Solid carriers for improved delivery of active ingredients in pharmaceutical compositions
US6446242B1 (en) * 1999-04-02 2002-09-03 Actel Corporation Method and apparatus for storing a validation number in a field-programmable gate array
US6635249B1 (en) 1999-04-23 2003-10-21 Cenes Pharmaceuticals, Inc. Methods for treating congestive heart failure
CN1138785C (en) * 1999-06-04 2004-02-18 周明东 Application of growth factor neuregulin and its analogs
US6458387B1 (en) 1999-10-18 2002-10-01 Epic Therapeutics, Inc. Sustained release microspheres
US6352721B1 (en) * 2000-01-14 2002-03-05 Osmotica Corp. Combined diffusion/osmotic pumping drug delivery system
ES2610353T3 (en) 2000-05-23 2017-04-27 Cenes Pharmaceuticals, Inc. NRG-2 nucleic acid molecules, polypeptides and diagnostic and therapeutic methods
EP1296660A4 (en) 2000-06-27 2005-04-13 Mi Tech Company Ltd The controlled release preparation of insulin and its method
WO2002013786A2 (en) 2000-08-15 2002-02-21 Board Of Trustees Of The University Of Illinois Method of forming microparticles
US6866866B1 (en) 2000-11-03 2005-03-15 Andrx Labs, Llc Controlled release metformin compositions
US6436386B1 (en) 2000-11-14 2002-08-20 Shearwater Corporation Hydroxyapatite-targeting poly (ethylene glycol) and related polymers
IN190699B (en) 2001-02-02 2003-08-16 Sun Pharmaceutical Ind Ltd
JP2004528345A (en) * 2001-04-30 2004-09-16 シャイア ラボラトリーズ,インコーポレイテッド Pharmaceutical composition comprising an ACE / NEP inhibitor and a bioavailability enhancer
CN1314445C (en) 2001-05-21 2007-05-09 耐科塔医药公司 Pulmonary administration of chemically modified insulin
US6838093B2 (en) 2001-06-01 2005-01-04 Shire Laboratories, Inc. System for osmotic delivery of pharmaceutically active agents
CN100424175C (en) 2002-03-26 2008-10-08 上海泽生科技开发有限公司 Methods and compositions for treating neoplasms by ERBB3
AU2002304965A1 (en) * 2002-05-24 2003-12-12 Zensun (Shanghai) Sci-Tech.Ltd Neuregulin based methods and compositions for treating viral myocarditis and dilated cardiomyopathy
CN1498656A (en) * 2002-11-08 2004-05-26 上海泽生科技开发有限公司 Method and compsn. of nervous regulation protein for treating myocardial infarction
US20070213264A1 (en) * 2005-12-02 2007-09-13 Mingdong Zhou Neuregulin variants and methods of screening and using thereof
EP1981525B1 (en) 2005-12-30 2015-01-21 Zensun (Shanghai) Science and Technology Limited Extended release of neuregulin for improved cardiac function
US9580515B2 (en) 2006-08-21 2017-02-28 Zensun (Shanghai) Science & Technology, Co., Ltd. Neukinase, a downstream protein of neuregulin
JP4616237B2 (en) 2006-11-07 2011-01-19 日本電信電話株式会社 Method for forming silicon compound thin film
US20090156488A1 (en) 2007-09-12 2009-06-18 Zensun (Shanghai) Science & Technology Limited Use of neuregulin for organ preservation
CN102231987A (en) 2008-11-28 2011-11-02 上海泽生科技开发有限公司 Neuregulin and cardiac stem cells
CN102232084B (en) 2008-11-28 2014-05-28 上海泽生科技开发有限公司 Neuregulin peptides and their use
ES2748886T3 (en) 2009-06-09 2020-03-18 Zensun Shanghai Science & Tech Co Ltd Neuregulin-based methods for the treatment of heart failure
WO2010142141A1 (en) 2009-06-09 2010-12-16 Zensun (Shanghai) Science & Technology Limited Neuregulin based methods for treating heart failure
JP6096262B2 (en) 2009-08-25 2017-03-15 ゼンサン (シャンハイ) サイエンス アンド テクノロジー,シーオー.,エルティーディー. Treatment of heart failure based on neuregulin
CN102139095A (en) 2010-01-29 2011-08-03 上海泽生科技开发有限公司 Method for applying neuregulin to prevent, treat or delay cardiac ischemia reperfusion injuries, and composition for preventing, treating or delaying cardiac ischemia reperfusion injuries
WO2013053076A1 (en) 2011-10-10 2013-04-18 Zensun (Shanghai)Science & Technology Limited Compositions and methods for treating heart failure
BR112015029293A2 (en) 2013-05-22 2018-04-24 Zensun Shanghai Science & Tech Ltd method and kit for preventing, treating or delaying a cardiovascular disease or disorder in a mammal
CN110946993A (en) 2014-01-03 2020-04-03 上海泽生科技开发股份有限公司 Formula of neuregulin preparation
CN105497876B (en) 2014-09-24 2021-01-15 上海泽生科技开发股份有限公司 Methods and compositions for the prevention, treatment or delay of cardiac ventricular arrhythmias with neuregulin
CN105561298A (en) 2014-10-17 2016-05-11 上海泽生科技开发有限公司 Method for preventing, treating or delaying ejection fraction reserved cardiac failure by means of neuregulin and composition

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
J. J. M. MEYEL ET AL: "Continuous infusion of furosemide in the treatment of patients with congestive heart failure and diuretic resistance", JOURNAL OF INTERNAL MEDICINE, vol. 235, no. 4, 1 April 1994 (1994-04-01), pages 329 - 334, XP055091961, ISSN: 0954-6820, DOI: 10.1111/j.1365-2796.1994.tb01082.x *
SINDONE ET AL: "Continuous home ambulatory intravenous inotropic drug therapy in severe heart failure: Safety and cost efficacy", AMERICAN HEART JOURNAL, MOSBY- YEAR BOOK INC, US, vol. 134, no. 5, 1 November 1997 (1997-11-01), pages 889 - 900, XP005926872, ISSN: 0002-8703, DOI: 10.1016/S0002-8703(97)80012-9 *

Also Published As

Publication number Publication date
JP5738516B2 (en) 2015-06-24
EP2918283B1 (en) 2018-01-31
CA2634974A1 (en) 2007-07-12
RU2457854C2 (en) 2012-08-10
EP1981525A4 (en) 2009-11-11
US20160095903A1 (en) 2016-04-07
EP2918283A1 (en) 2015-09-16
JP5948306B2 (en) 2016-07-06
ES2530526T3 (en) 2015-03-03
AU2006332340B2 (en) 2013-09-26
JP2015199757A (en) 2015-11-12
BRPI0620819A2 (en) 2011-11-22
US20240075103A1 (en) 2024-03-07
JP2009522209A (en) 2009-06-11
RU2008131300A (en) 2010-02-10
US11638746B2 (en) 2023-05-02
JP2014122214A (en) 2014-07-03
EP3363455A1 (en) 2018-08-22
ES2664086T3 (en) 2018-04-18
WO2007076701A1 (en) 2007-07-12
EP1981525A1 (en) 2008-10-22
US20070190127A1 (en) 2007-08-16
AU2006332340A1 (en) 2007-07-12
CA2841386A1 (en) 2007-07-12

Similar Documents

Publication Publication Date Title
US20240075103A1 (en) Extended release of neuregulin for improved cardiac function
JP7100083B2 (en) Prescription of Neuregulin formulation
Liang et al. The use of folate-PEG-grafted-hybranched-PEI nonviral vector for the inhibition of glioma growth in the rat
Matsumura et al. Phase I and pharmacokinetic study of MCC-465, a doxorubicin (DXR) encapsulated in PEG immunoliposome, in patients with metastatic stomach cancer
US8173170B2 (en) Drug carrier and drug carrier kit for inhibiting fibrosis
CN101394861A (en) Extended release of neuregulin for improved cardiac function
JP2022023208A (en) Therapeutic dosing of neuregulin or subsequence thereof for treatment or prophylaxis of heart failure
US20090162425A1 (en) Methods and compositions for inhibiting undesirable cellular proliferation by targeted liposome delivery of active agents
CA3008015A1 (en) Controlled-release c-type natriuretic peptide agonists with increased neutral endopeptidase stability
EP3270889B1 (en) Targeted liposomal delivery of cgmp analogues
CA3008017A1 (en) Controlled-release cnp agonists with reduced side-effects
KR102444612B1 (en) Formulations comprising recombinant acid alpha-glucosidase
AU2013203483B2 (en) Extended release of neuregulin for improved cardiac function
Guo et al. Pre-clinical efficacy and safety pharmacology of PEGylated recombinant human endostatin
CN103768582A (en) Neuregulin capable of improving heat function when continually administrated

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20080728

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LI LT LU LV MC NL PL PT RO SE SI SK TR

AX Request for extension of the european patent

Extension state: AL BA HR MK RS

A4 Supplementary search report drawn up and despatched

Effective date: 20090911

17Q First examination report despatched

Effective date: 20091204

RAP1 Party data changed (applicant data changed or rights of an application transferred)

Owner name: ZENSUN (SHANGHAI) SCIENCE AND TECHNOLOGY LIMITED

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

INTG Intention to grant announced

Effective date: 20141110

GRAS Grant fee paid

Free format text: ORIGINAL CODE: EPIDOSNIGR3

GRAA (expected) grant

Free format text: ORIGINAL CODE: 0009210

AK Designated contracting states

Kind code of ref document: B1

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LI LT LU LV MC NL PL PT RO SE SI SK TR

AX Request for extension of the european patent

Extension state: AL BA HR MK RS

REG Reference to a national code

Ref country code: GB

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: CH

Ref legal event code: EP

REG Reference to a national code

Ref country code: IE

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: CH

Ref legal event code: NV

Representative=s name: KIRKER AND CIE S.A., CH

REG Reference to a national code

Ref country code: AT

Ref legal event code: REF

Ref document number: 707633

Country of ref document: AT

Kind code of ref document: T

Effective date: 20150215

REG Reference to a national code

Ref country code: ES

Ref legal event code: FG2A

Ref document number: 2530526

Country of ref document: ES

Kind code of ref document: T3

Effective date: 20150303

REG Reference to a national code

Ref country code: DE

Ref legal event code: R096

Ref document number: 602006044395

Country of ref document: DE

Effective date: 20150305

REG Reference to a national code

Ref country code: SE

Ref legal event code: TRGR

REG Reference to a national code

Ref country code: NL

Ref legal event code: T3

REG Reference to a national code

Ref country code: AT

Ref legal event code: MK05

Ref document number: 707633

Country of ref document: AT

Kind code of ref document: T

Effective date: 20150121

REG Reference to a national code

Ref country code: LT

Ref legal event code: MG4D

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: FI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150121

Ref country code: BG

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150421

Ref country code: LT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150121

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: AT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150121

Ref country code: IS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150521

Ref country code: PL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150121

Ref country code: LV

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150121

Ref country code: GR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150422

REG Reference to a national code

Ref country code: DE

Ref legal event code: R097

Ref document number: 602006044395

Country of ref document: DE

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150121

Ref country code: EE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150121

Ref country code: RO

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150121

Ref country code: DK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150121

Ref country code: CZ

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150121

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 10

PLBE No opposition filed within time limit

Free format text: ORIGINAL CODE: 0009261

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: NO OPPOSITION FILED WITHIN TIME LIMIT

26N No opposition filed

Effective date: 20151022

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150121

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: BE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150121

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LU

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20151229

Ref country code: MC

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150121

REG Reference to a national code

Ref country code: IE

Ref legal event code: MM4A

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: IE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20151229

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 11

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: HU

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT; INVALID AB INITIO

Effective date: 20061229

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: CY

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150121

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: TR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150121

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 12

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: PT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150121

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: ES

Payment date: 20230112

Year of fee payment: 17

Ref country code: CH

Payment date: 20230101

Year of fee payment: 17

P01 Opt-out of the competence of the unified patent court (upc) registered

Effective date: 20230527

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: NL

Payment date: 20231116

Year of fee payment: 18

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: GB

Payment date: 20231109

Year of fee payment: 18

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: SE

Payment date: 20231110

Year of fee payment: 18

Ref country code: IT

Payment date: 20231110

Year of fee payment: 18

Ref country code: FR

Payment date: 20231108

Year of fee payment: 18

Ref country code: DE

Payment date: 20231031

Year of fee payment: 18

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: ES

Payment date: 20240115

Year of fee payment: 18